Skip to main content

REVIEW article

Front. Genet., 30 November 2021
Sec. Computational Genomics
This article is part of the Research Topic Methods and Applications: Computational Genomics View all 43 articles

Current Progress and Future Perspectives of Immune Checkpoint in Cancer and Infectious Diseases

Xin Cai&#x;Xin Cai1Huajie Zhan&#x;Huajie Zhan2Yuguang Ye&#x;Yuguang Ye3Jinjin YangJinjin Yang2Minghui Zhang
Minghui Zhang4*Jing Li
Jing Li5*Yuan Zhuang
&#x;Yuan Zhuang2*
  • 1Heilongjiang Administration of Traditional Chinese Medicine, Harbin, China
  • 2Department of Pathology, Harbin Medical University, Harbin, China
  • 3Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, China
  • 4Department of Oncology, Chifeng City Hospital, Chifeng, China
  • 5Department of Pathology and Electron Microscopy Center, Harbin Medical University, Harbin, China

The inhibitory regulators, known as immune checkpoints, prevent overreaction of the immune system, avoid normal tissue damage, and maintain immune homeostasis during the antimicrobial or antiviral immune response. Unfortunately, cancer cells can mimic the ligands of immune checkpoints to evade immune surveillance. Application of immune checkpoint blockade can help dampen the ligands expressed on cancer cells, reverse the exhaustion status of effector T cells, and reinvigorate the antitumor function. Here, we briefly introduce the structure, expression, signaling pathway, and targeted drugs of several inhibitory immune checkpoints (PD-1/PD-L1, CTLA-4, TIM-3, LAG-3, VISTA, and IDO1). And we summarize the application of immune checkpoint inhibitors in tumors, such as single agent and combination therapy and adverse reactions. At the same time, we further discussed the correlation between immune checkpoints and microorganisms and the role of immune checkpoints in microbial-infection diseases. This review focused on the current knowledge about the role of the immune checkpoints will help in applying immune checkpoints for clinical therapy of cancer and other diseases.

Introduction

Activation of T cells plays an important role in the process of immunity (Lenschow and Bluestone, 1993). During normal immune response, the process that T cells accept antigen peptides presented by major histocompatibility complex (MHC) on antigen-presenting cells (APCs) via T-cell receptor (TCR) in order to exert its function is called the first signal for T-cell activation. The second signal for T-cell activation is a costimulatory signal which comes from a combination between CD28 on T cells and CD80(B7-1)/CD86(B7-2) on APCs (Lenschow et al., 1996; Nandi et al., 2020). This activation process also requires cytokines such as IL-2 to help. The rightly activated T cells or in tandem with B cells will eliminate threats, while uncontrolled activation of T cells would bring serious consequences such as autoimmune diseases (Takeuchi et al., 2020). Therefore, scientists devoted their lives to shed light on how the immune system regulates itself.

In the last two decades, the understanding of regulatory pathways in immune responses to cancer immunotherapies remains unclear. The enormous progress was made in 1996; Leach and his colleagues (Linsley et al., 1991; Leach et al., 1996) have been validated that blockade of cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) could downregulate T-cell responses and enhance antitumor responses in immunocompetent mouse models. In 2000, Gordon J. Freeman identified that CTLA-4 structurally similar protein-programmed death 1 (PD-1) could bind to its ligand PD-L1 and lead to the inhibition of lymphocyte proliferation (Freeman et al., 2000). The binding of B- and T-cell lymphocyte attenuator (BTLA) to its ligand HVEM may lead to decreased T-cell proliferation and cytokine production (Murphy et al., 2006). The binding of T-cell immunoglobulin and mucin domain-containing 3 (TIM-3) to its ligand galectin-9 could result in T helper 1 (Th1) cell death (Zhu et al., 2005). V-domain Ig suppressor of T-cell activation (VISTA) is a potent T-cell suppressor and inhibits T-cell immune response in animal models (Wang et al., 2011). During these processes, the set of costimulatory or coinhibitory molecules, which regulate the activation, effector functions, and interactions among APCs and T lymphocytes, provides a critical checkpoint in the regulation of T-cell immunity and maintenance of immune homeostasis. As their function in the balance of the immune system, these costimulatory or coinhibitory proteins are defined as immune checkpoint proteins (Figure 1, Table 1). A direct consequence of these findings was to reveal the regulatory pathways involved in immune responses in cancer and infectious diseases.

FIGURE 1
www.frontiersin.org

FIGURE 1. Immune checkpoint receptors and their ligands. Two signals participate in T-cell activation: 1) T cells recognize antigen presented by MHC-II molecules on APCs through TCR; 2) T cells accept costimulatory signals CD80/CD86 through CD28.

TABLE 1
www.frontiersin.org

TABLE 1. The expression and mechanism of the immune checkpoints.

Immune checkpoint proteins have been playing a significant role in inflammatory reactions and cancer immunotherapy. A number of immune checkpoint proteins were shown to be dysregulated in cancers and infectious diseases, including PD-1/PD-L1, CTLA-4, lymphocyte activation 3 (LAG-3), TIM-3, VISTA, and Indoleamine-2,3 dioxygenase 1 (IDO1). These immune checkpoints and other regulatory cells, such as regulatory T cells (Tregs), myeloid-derived suppressors cells (MDSCs), M2 macrophages, and cytokines, are often enhanced during infections and cancers (Pauken and Wherry, 2015). Pathogens can develop immune checkpoints to limit host-protective antigen-specific immune response (Dyck and Mills, 2017). The cancer cells can disrupt the immune response and cleverly escape from immunity by dysregulating immune checkpoint signaling. Many similarities exist between cancer and infectious disease (Hotchkiss and Moldawer, 2014). They can utilize similar receptors to detect damage-associated molecular patterns (DAMP) and pathogen-associated molecular patterns (PAMPs), respectively (Vance et al., 2017). In the meantime, persistent stimulation of the immune system and induction of T-cell-mediated inflammation can be aroused. In pathogen-infected diseases, with elevated expression of the immune checkpoint molecules on T cells as it is in cancer, the immune checkpoint blockade therapy may bring favorable consequences (Wykes and Lewin, 2018). So, agonists of costimulatory signals or antagonists of inhibitory signals function as good ways for cancer therapy and also could help to reverse the state of immune suppression in chronic infection. Some antibodies that targeted immune checkpoint molecules to reverse the suppression of the immune system have been applied in the clinical treatment of cancer (Remon and Besse, 2017; Chen et al., 2019). However, the unexpected events of an immune checkpoint inhibitor (ICI) have emerged as frequent complications at the same time.

Here, we review the mechanisms, functions, and adverse events of common immune checkpoints in cancer and infectious diseases. We also discuss the impact of the bacterial microbiome on the relationship between cancer therapy and the immune system.

Biology of Immune Checkpoint Proteins

PD-1/PD-L1

PD-1 is a 288 amino acid protein that is encoded by the PDCD1 gene and belongs to the immunoglobulin superfamily (Tavares et al., 2018). PD-1 can be expressed on T cells, B cells, natural killer cells (NKs), dendritic cells (DCs), macrophages, and monocytes (Ahmadzadeh et al., 2009). T cells inducibly express PD-1 after activation (Han et al., 2020), while different from other members of the CD28 superfamily, which has Src homology (SH2) binding motifs and/or SH3 binding motifs in their cytoplasmic tail, the cytoplasmic tail of PD-1 possesses a sequence that can form an immunoreceptor tyrosine-based inhibition motif (ITIM) and an immunoreceptor tyrosine-based switch motif (ITSM) that can recruit Src homology 2 domain-containing protein tyrosine phosphatases (SHP-2), resulting in the inhibitory function (Neel et al., 2003; Patsoukis et al., 2020).

The two ligands of PD-1, PD-L1 (also known as B7-H1) and PD-L2 (B7-H2), differ in expression patterns (Panjwani et al., 2018). PD-L1 is expressed on many cells, including B cells, T cells, macrophages, tumor cells, and other tissue cells such as vascular endothelial cells (Ritprajak and Azuma, 2015; Dermani et al., 2019). Ligation of PD-1 and PD-L1 can lead to T-cell dysfunction and anergy, helping PD-L1 expressing tumor cells escape from cytotoxic T-cell-mediated cell death (Ritprajak and Azuma, 2015; Dermani et al., 2019).

PD-1/PD-L1 blockade not only facilitates T-cell function but also restores NKs antitumor response (Hsu et al., 2018). PD-L1 expression on cancer cells resulted in the generation of more aggressive tumors in vivo. Depleting NKs before PD-L1 expressed or not tumor cell implantation resulted in similar growth of tumors and mortality. However, no such effect occurring with depletion of CD4+ and CD8+ T cells indicates that NKs take a vital position in immune checkpoint blockade (Hsu et al., 2018).

It is reported that several signaling pathways would participate in the PD-1/PD-L1 axis. For example, PD-1PD-L1+ regulatory B cells must exert their immunosuppressive function through activation of the PI3K/AKT/NF-κB signaling pathway in breast cancer (Liu et al., 2021). PTEN is a critical inhibitor of the PI3K/AKT signaling pathway. In microsatellite instability-high (MSI-H) or mismatch repair deficient (dMMR) gastrointestinal tumors, mutation of PTEN, especially in the phosphatase domain, could be negative predictors of PD-1 blockade treatment (Chida et al., 2021). Blockade of MAPK pathway through MEK1 and two inhibitors prevented the expression of PD-L1 in lung adenocarcinoma cells (Stutvoet et al., 2019), whereas inhibition of ERK could improve the anti-PD-L1 checkpoint blockade effect in preclinical pancreatic ductal adenocarcinoma (Henry et al., 2021). What we have listed above indicates that MAPK pathway activity could also severely influence the PD-L1 axis despite the PI3K pathway. Similarly, using inhibitors of the JAK/STAT pathway, which was reported to suppress PD-L1 upregulation, showed that it can also take part in regulating the PD-L1 axis (Doi et al., 2017).

CTLA-4

CTLA-4 is a 223 amino acid protein, which belongs to the immunoglobulin superfamily and consists of an IgV domain, a transmembrane region, and a cytoplasmic tail containing a conserved YVKM motif (Rowshanravan et al., 2018). Stored in endocytic vesicles, CTLA-4 is transported to the cell membrane to be colocalized with TCR on the cell surface. Dependent on dynamin and clathrin adaptor protein complex (AP2), which targets the YVKM motif, internalization of CTLA-4 from cell surface for degradation and recycling is rapid (usually within minutes) (Shiratori et al., 1997). Then CTLA-4 can be transported to cell membrane again or compartment of lysosome for degradation. Such regulation of AP2 can be disrupted by the phosphorylation of the YVKM motif after T-cell activation (Qureshi et al., 2012). Lipopolysaccharide responsive and beige-like protein (LRBA) may inhibit degradation of CTLA-4 by disrupting transportation of CTLA-4 to the lysosome via binding to YVKM sequence and promote recycling of CTLA-4. Patients with LRBA deficiency raised autoimmunity syndrome designating that accurate CTLA-4 trafficking is important for autoimmune diseases (Lo et al., 2015; Rowshanravan et al., 2018).

The phenomenon that CTLA-4, often expressed on antigen-specific T cells, has a higher affinity (10–100-fold) for CD80 dimer and CD86 monomer than CD28 is considered to be a conventional concept about how CTLA-4 downregulates the immune response (Linsley et al., 1994; van der Merwe et al., 1997). Different from antigen-specific T cells that upregulated CTLA-4 after activation, Tregs constitutively express a high range of CTLA-4 ensuring immune homeostasis and immunosuppressive capacity. Intriguingly, there have been studies proved that CD80 and CD86 on APC can be captured and deleted by CTLA-4 expressed on CD4+CD25+Foxp3+ Tregs (Qureshi et al., 2011; Tekguc et al., 2021), while patients or carriers with CTLA-4 mutation showed diminished Tregs inhibitory function and impaired trans-endocytosis of CD80 (Schubert et al., 2014). These discoveries provide a proper explanation for the rapid endocytic behavior of CTLA-4 that CTLA-4 may exhibit its inhibitory function by trans-endocytosis. Also, there have been studies about other mechanisms undergoing CTLA-4 inhibition. Kong et al. found that protein kinase C-η (PKC-η) was recruited to and physically associated with the CTLA-4 expressed on Tregs in the immunological synapse. PKC-η-deficient Tregs lacked their suppressive function, leading to lymphoproliferation and autoimmune syndromes (Kong et al., 2014). In addition, competitively binding with CD28, CTLA-4 limited the positive costimulation of CD28 by blocking the downstream PI3K/AKT and NF-κB signaling pathway (Pages et al., 1994; Olsson et al., 1999). The anti-CTLA-4 antibody (ipilimumab) eliminated Tregs in an Fc-dependent manner to achieve clinical relief, which may be due to relieved NKs cytotoxicity suppressed by Tregs (Romano et al., 2015; Khan et al., 2020). For anti-CTLA-4 antibodies therapy, CD8+ T cells were required for the therapeutic effect. Fas-FasL and perforin interactions also were important for CTLA-4 blockade (van Elsas et al., 2001).

LAG-3

Firstly identified in 1990 by Triebel and colleagues, lymphocyte activation 3 (LAG-3, CD223), an immune inhibitory receptor, is a 503 amino acid protein encoded by lymphocyte activation gene that is located on chromosome 12, containing eight exons (Triebel et al., 1990; Sierro et al., 2011). Belonging to the Ig superfamily, LAG-3 contains four extracellular Ig-like domains D1, D2, D3, and D4, which share approximately 20% amino acid homology with that of CD4. Comprising unlike intracellular region with CD4, LAG-3 is closely related but exhibits divergent functions with CD4 (Maruhashi et al., 2020). The cytoplasmic tail of LAG-3 has three conserved motifs. The first motif, which has not been considered functional, contains a hypothesized serine phosphorylation site containing two serine residues in humans. It is reported that the second motif, which has conserved six amino acid sequences (KIEELE), plays an important role in dampening T-cell proliferation, cytokine production, and cytolytic function. The third motif is a glutamic acid and proline dipeptide repeat which can colocalize LAG-3 with CD3, CD4, and CD8 molecules (Goldberg and Drake, 2011; Ruffo et al., 2019).

LAG-3 can be detected from CD4+ and CD8+ T cells, Tregs, NKs, and plasmacytoid DCs and do not express on naive T cells similar to PD-1 and CTLA-4 (Goldberg and Drake, 2011). Activation of LAG-3 can elevate intratumoral Tregs activity, and blocking of it will upregulate T-cell function and reinvigorate CD8+ tumor-infiltrating lymphocytes (TILs) to eliminate tumor cells (Lecocq et al., 2020). CD4+CD25+ Tregs from LAG-3 (−/−) mice exhibited reduced regulatory activity. Treated with anti-LAG-3 antibody, suppression induced by Tregs was inhibited in vitro and in vivo. It is obvious that LAG-3 marks Tregs populations and intermediates their regulatory function (Huang et al., 2004). As a transmembrane protein receptor which is similar to CD4 with greater affinity for MHC-II molecules on APCs (Triebel et al., 1990), there are also other proposed ligands for LAG-3 like galectin-3, fibrinogen-like protein 1 (FGL-1), α-synuclein, and LSECtin (Xu et al., 2014; Kouo et al., 2015; Mao et al., 2016). Recent research showed that FGL-1 worked as an important ligand of LAG-3 in its inhibitory effect on T cells. The expression of LAG-3 can be elevated on exhausted T cells in cancer. FGL-1 is upregulated in several human cancers, and genetic ablation or blockade of the FGL-1/LAG-3 interaction by monoclonal antibodies (mAbs) would enhance T-cell responses and antitumor immunity. Wang et al. expected a poor prognosis in non-small-cell lung cancer (NSCLC) patients with high plasma FGL-1 treated with anti-PD therapy (Wang et al., 2019a). The precise function of ligands of LAG-3 still needs to be clarified.

TIM-3

TIM-3 is a transmembrane protein encoded by HAVCR2 and identified on IFN-γ-producing CD4+ Th1 cells and CD8+ type 1 cytotoxic T cells firstly. Then it is also discovered on monocytes, Tregs, DCs, and NKs (Wolf et al., 2020). The fact that administration of antibody to TIM-3 could enhance Th1-dependent autoimmune disease strongly implying that TIM-3 works as an inhibitory molecule on T-cell function (Monney et al., 2002). Indeed, TIM-3 is found to be coregulated and coexpressed with other immune checkpoint receptors, such as PD-1 and LAG-3 (Chihara et al., 2018). High expression of TIM-3 on effector T cells also indicates severe T-cell exhaustion or dysfunction (Avery et al., 2018).

Without known inhibitory signaling motifs in its cytoplasmic tail, TIM-3 contains five conserved tyrosines to play its role. TIM-3 can be found in lipid rafts and is recruited to the immunological synapse upon T-cell activation (Clayton et al., 2014). TIM-3 interacts with HLA-B associated transcript (BAT3) in ligand unbound form and maintains T-cell activation by recruiting an active form of tyrosine kinase LCK, while in ligand-bound form, tyrosine phosphorylation in its cytoplasmic tail will release BAT from TIM-3 and recruit tyrosine kinase FYN resulting in immune synapse disruption, phosphatase recruitment, and cell apoptosis (van de Weyer et al., 2006; Rangachari et al., 2012).

It has been demonstrated that IL-27/NFIL3 axis promotes permissive chromatin remodeling of the TIM-3 locus, induces TIM-3 expression, and is crucial for the induction of TIM-3 in vivo. IL-27-conditioned Th1 cells exhibit inhibitory function through NFIL3 in intestinal inflammation (Zhu et al., 2015). In human acute myeloid leukemia (AML), activation of TIM-3 works through NF-κB and β-catenin signaling pathways to promote self-renewal of leukemic stem cells (Kikushige et al., 2015). In hepatocellular carcinoma (HCC), TIM-3 was significantly upregulated in NKs and suppressed their cytokine production and cytotoxic activity through inhibiting PI3k/Akt/mTORC1 signaling pathway (Tan et al., 2020).

Different ligands of TIM-3 show various effects. The well-studied ligands of TIM-3 are galectin-9 (Gal-9), carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM-1), high mobility group box-1 protein (HMGB1), and phosphatidylserine (PtdSer). In T cells, ligation between Gal-9 and carbohydrate motifs on the IgV domain of TIM-3 functions in an immunosuppressive way which will induce T-cell apoptosis (Du et al., 2017; Dixon et al., 2018). CEACAM-1 coexpressed with TIM-3 is considered to be required for the regulatory function of TIM-3 (Huang et al., 2015). HMGB1 can bind to DNA released from dying cells and facilitate the uptake of DNA by Toll-like receptors. The interaction between HMGB1 and TIM-3 interferes with the innate immune response induced by nucleic acid (Nogueira-Machado et al., 2011; Chiba et al., 2012; Urban-Wojciuk et al., 2019). PtdSer-TIM-3 interaction shows clues for participating in apoptotic clearance cells, and more consequences between their interaction are waiting to be found (Nakayama et al., 2009).

Gal-9 binding with TIM-3 can cause an influx of calcium and mediate aggregation and apoptosis of effector Th1 cells in vitro. Administration of Gal-9 can result in selective loss of IFN-γ-producing cells and suppression of Th1 autoimmunity (Zhu et al., 2005). PtdSer engagement will induce TIM-3 phosphorylation leading to dysfunction of NKs in HCC (Tan et al., 2020). In head and neck squamous cell cancer (HNSCC), blockade of TIM-3 by mAbs induced the reduction of Tregs and increased IFN-γ production of CD8+ T cells, while the population of CD206+ M2 macrophages was not significantly reduced (Liu et al., 2018). Intriguingly, TIM-3 can also play an immunostimulatory role in NKs, DCs, and macrophages (Gleason et al., 2012; Zhang et al., 2012; Yang et al., 2013; Clayton et al., 2014).

VISTA

V-domain Ig suppressor of T cell activation (VISTA), also termed as PD-1H, B7-H5, V-set immunoregulatory receptor (VSIR), stress-induced secreted protein 1 (SISPQ), and differentiation of embryonic stem cells 1 (Dies1), is a conventional transmembrane protein whose IgV domain homology with PD-L1 and encoded by the gene located on chromosome 10 (Huang et al., 2020). Although containing a similar molecular sequence with the B7 superfamily, VISTA does not possess ITIM/ITAM (immunoreceptor tyrosine-based activation motif). VISTA is expressed on myeloid cells (e.g., monocytes, conventional DCs, macrophages, and circulating granulocytes), T cells, Tregs, and TILs (Hosseinkhani et al., 2021). There are increasing pieces of evidence showing VISTA as a regulatory immune checkpoint. In mice lacking VISTA, they would develop spontaneous T-cell activation, cutaneous lupus erythematosus, and production of inflammatory cytokines and chemokines (Wang et al., 2014; Liu et al., 2015; Han et al., 2019). With the presence of VISTA on erythroid cells, the transformation from naive CD4+ T cells to Tregs would be accelerated through the production of TGF-β (Shahbaz et al., 2018).

Though the binding pattern of VISTA is not clear, several studies showed that VISTA could act as both ligand on APCs and receptor on T cells (Flies et al., 2014; Lines et al., 2014). Researches have reported V-Set and Immunoglobulin domain containing 3 (VSIG-3) as the ligand for VISTA in impeding cytokine and chemokine production (Wang et al., 2019b). In consideration of elevated expression of VISTA or VSIG-3 in many cancers, such as colorectal cancer (CRC), HCC, and intestinal-type gastric cancers, the blockade of the VISTA/VSIG-3 pathway can work as a new target for immune checkpoint therapy. Besides, Alan et al. presented that VISTA can bind to P-selectin glycoprotein ligand-1 (PSGL-1) in a pH-dependent model (Johnston et al., 2019). Meanwhile, a study of VISTA in malignant pleural mesothelioma shows that VISTA expression was associated with better overall survival (OS), suggesting VISTA’s prognostic value (Muller et al., 2020).

IDO1

Indoleamine-2,3 dioxygenase 1 (IDO1) is one of the three enzymes which catalyze the first rate-limiting step in the oxidative metabolism of tryptophan, an essential amino acid for T-cell proliferation and differentiation. It is mainly distributed in DCs, macrophages, and monocytes (Munn and Mellor, 2013).

Tumor cells can recruit IDO-expressed DCs into the tumor microenvironment (TME). Due to the aggregation of IDO, lack of tryptophan will lead to stagnation of T-cell proliferation and differentiation in many ways. First, decreased tryptophan means elevated uncharged Trp-tRNA, which leads to activation of a stress response kinase, general control nonderepressible 2 (GCN2) (Munn et al., 2005). Then eukaryotic initiation factor-2 (eIF-2) is phosphorylated by GCN2, and translation of protein required for generation and proliferation of effector T cells will be limited. Second, degradation of tryptophan results in suppression of mammalian target of rapamycin complex 1 (mTORC1) and PKC-θ associated with induction of autophagy. Apoptosis of effector T cells will be reinforced (Metz et al., 2012). Third, IDO1 can induce Tregs through increased activity of aryl hydrocarbon receptor (AHR) binding with kynurenine, a metabolite of tryptophan (Mezrich et al., 2010). Thus, the unbalanced metabolism of tryptophan can promote tumor development and evade immune detection indicating that the application of IDO1 inhibitor is also a promising means to enhance antitumor immunity in theory. In status quo, clinical application of IDO1 inhibitor displayed a controversial outcome with rare effect on monotherapy and combination therapy. Although the agents might not be suitable for such types of cancer involved in research, they may be helpful in other diseases.

Single Agent and Combined Therapy in Cancer

Balckburn et al. have demonstrated that T-cell function decreases with increased expression of immune checkpoints, so targeting these immune checkpoint proteins to modulate immune responses holds great promise for cancer immunotherapy (Blackburn et al., 2009). The purpose of immune checkpoint blockade is mainly to suppress CD8+ T cells and improve tumor-specific immune response. The mAbs by targeting checkpoints CTLA-4 and PD-1/PD-L1 have achieved the US Food and Drug Administration (FDA) approval for the treatment of different cancers (Peggs et al., 2006; Hodi et al., 2010).

Ipilimumab was the first FDA-approved recombinant humanized anti-CTLA-4 immunoglobulin G1 monoclonal antibody in 2011 for the treatment of advanced melanoma in patients who cannot be surgically cured or have metastasis (Vaddepally et al., 2020). It can also work well with intermediate or poor-risk advanced renal cell carcinoma (RCC), MSI-H/dMMR CRC, metastatic NSCLC, unresectable malignant pleural mesothelioma, and HCC, which have been previously treated with sorafenib, in combination with nivolumab (Pinto et al., 2019; McKay et al., 2020; Baas et al., 2021; Casak et al., 2021; Saung et al., 2021). In 2014, nivolumab and pembrolizumab (PD-1 blockade) were approved by the FDA as a humanized IgG antibody for the treatment of unresectable or metastatic melanoma (Prasad and Kaestner, 2017; Finkelmeier et al., 2018). In 2016, the PD-L1 blockade, atezolizumab, a humanized IgG antibody, officially worked as a second-line treatment for locally advanced or metastatic urothelial carcinoma (Patel et al., 2017). With the maturity of theory and technology, the usage range of PD-1/PD-L1 blockade has gradually expanded, including metastatic nonsquamous NSCLC, advanced RCC, unresectable or metastatic, recurrent HNSCC, MSI-H/dMMR CRC, relapsed or refractory classical Hodgkin lymphoma (cHL), locally advanced or metastatic urothelial carcinoma, cervical cancer, gastric cancer, and esophageal cancer (Ansell et al., 2015; Beckermann et al., 2017; Chae et al., 2018; Lin et al., 2018; Saito et al., 2018; Oliveira et al., 2019; Wang and Li, 2019; Nassar et al., 2020; Wu et al., 2020). Pembrolizumab and nivolumab targeting PD-1 showed promising results in melanoma and NSCLC with an objective response rate (ORR) of 40–45% (Darvin et al., 2018). LAG-3 is coexpressed with many inhibitory immune checkpoints, especially PD-1, and this signifies a more exhausting state than expressing PD-1 alone. Utilization of coblockade for PD-1 and LAG-3 shows better curative effects. Relatlimab (in combination with nivolumab) is the first LAG-3 blocking antibody to demonstrate a benefit for patients in a Phase 3 study (Lipson et al., 2021). IMP321, a recombinant soluble LAG-3 Ig fusion protein of which multiple phases I and phase II trials have been completed, may enhance T-cell response, expand the percentage of long-lived effector-memory CD8+ T cells, and rarely induce immune-related adverse events (irAEs) (Brignone et al., 2009; Wang-Gillam et al., 2013). TIM-3, as an immunoinhibitory molecule, indicates the most terminal state of T cells, whose antibodies are being studied and evaluated for clinical trials, including Sym023 (NCT03489343), TSR-022 (NCT03680508), LY3321367 (NCT03099109), and MBG453 (NCT02608268). Many studies focus on the combination between anti-TIM-3 antibody and anti-PD-1 antibody in patients with advanced relapsed or a refractory solid tumor. There are also some ongoing clinical trials that evaluate the safety and feasibility of different ICIs in various tumors. Therapeutically targeting BTLA, VISTA, TIM-3, and TIGIT remain in preclinical stages to treat advanced solid malignancies (Derre et al., 2010) (NCT02671955, NCT02817633, NCT02608268, and NCT03119428).

The combination of immune checkpoints may improve clinical response rates. CTLA-4 and PD-1 blockade combination could increase effector T-cell infiltration into B16 melanoma in mice (Curran et al., 2010). Nivolumab plus ipilimumab in patients with metastatic melanoma yielded a response rate from 40% with treatment alone to 72% among patients who were PD-L1-positive (Larkin et al., 2015). In an open-label, randomized, phase 3 study (CheckMate 743), the results showed that nivolumab plus ipilimumab prolonged the median of the OS by nearly one-third versus chemotherapy (18.1 versus 14.1 months) and 2-years OS rates by nearly a half (41 versus 27%) (Baas et al., 2021). Early data using relatlimab plus nivolumab showed promising antitumor activity with an 11.5% ORR (NCT01968109). Now more and more researches focus on combination medication on relatlimab in HCC (NCT04658147), melanoma (NCT03743766), refractory MSI-H solid tumor (NCT03607890), HNSCC (NCT04326257), and so on. Although the clinical effectiveness of these ICIs gained great success in cancer immunotherapy, a subset of patients still does not respond to these inhibitors.

There are also some studies that showed that immune checkpoint blockade combined with radiotherapy, chemotherapy, and targeted drugs could improve the antitumor efficacy (Twyman-Saint Victor et al., 2015; Ebert et al., 2016; Shi et al., 2016). In the murine HCC model, combination with anti-TIM-3 and radiotherapy significantly shrink the tumor growth and elongate the OS compared with monotherapy (Kim et al., 2021). In an open-label, randomized, phase III trial (CheckMate 649), nivolumab plus chemotherapy reveals promising prospects than chemotherapy alone with superior OS and progression-free survival (PFS) benefit (Janjigian et al., 2021). Guidelines recommended using atezolizumab plus nab-paclitaxel for first-line treatment of unresectable, locally advanced, or metastatic triple-negative breast cancer (TNBC) with PD-L1 expressed on tumor-infiltrating immune cells. A survival analysis found that the OS, safety outcomes, and occurrence of immune-mediated adverse events of atezolizumab plus nab-paclitaxel were all ameliorated than placebo plus nab-paclitaxel (Emens et al., 2021). A TLR9 binding CpG-ODN adjuvant with a systemic anti-CTLA-4 antibody could increase the survival of mice bearing poorly immunogenic B16 melanoma (Davila et al., 2003).

Immune-Related Adverse Events Induced by ICIs

As we know, immune checkpoint blockade has demonstrated a significant promise in the clinic across a range of cancer indications (Chen and Mellman, 2017). However, the immune checkpoint blockade can reinforce host immunity at an expanse of uncontrolled effects that results in a unique spectrum of toxicities defined as immune-related adverse effects (irAEs) (Xu et al., 2018). The degree of irAEs is divided into five grades, comprising mild, moderate, severe, life-threatening, and death, elucidated on Common Terminology Criteria for adverse events from US National Cancer Institute (Cancer Therapy Evaluation Program, 2017). Some key oncology societies recently published comprehensive guidelines for irAEs, including the American Society of Clinical Oncology (ASCO), the European Society for Medical Oncology (ESMO), the Society for Immunotherapy of Cancer Toxicity Management Working Group, and the National Comprehensive Cancer Network (Connolly et al., 2019; Ramos-Casals et al., 2020). The referred organs/system of irAEs include, but are not limited to, cardiac, dermatological, endocrine, gastrointestinal, neurological, muscular, pulmonary, ocular, renal, skeletal, and systemic toxicities.

Paolo et al. declared that irAEs occurring in patients treated with ipilimumab were dose-dependent (Ascierto et al., 2017). Generically, the earliest and the most frequent symptom that showed up during ICI therapy (both anti-CTLA-4 and anti-PD-1) was dermatological changes (Sandigursky and Mor, 2018). A meta-analysis of irAEs in phase III randomized controlled trials of lung cancer proposed that the most frequent irAEs were diarrhea, skin rash, and hypothyroidism (Berti et al., 2021). Another network meta-analysis specifically presented that the main irAEs of ipilimumab were related to the gastrointestinal system (diarrhea, 29%) and skin (rash, 31%), while nivolumab and pembrolizumab were referred to as less frequency in irAEs with maculopapular rash (13%), erythema (4%), hepatitis (3%), arthralgia (12%), hypothyroidism (8%), and hyperglycemia (6%), respectively (Almutairi et al., 2020). A retrospective analysis about North American Intergroup trial E1609 with 1,673 patients proclaimed that grade 1-2 irAEs were associated with longer relapse-free survival (RFS) and OS versus no irAEs, while grade 3-4 showed lesser benefit from RFS and no benefit from OS (Tarhini et al., 2021). Combined immunotherapy could induce more severe and sustained irAEs than monotherapy (Choi and Lee, 2020).

T cells can undergo spontaneous differentiation into Tfh cells in CTLA-4-deficient mice, while not in CD28-deficient mice, they might be applied to explain lethal multiorgan autoimmune symptoms in CTLA4−/− mice (Walker, 2017). As precise mechanisms of irAEs have not been elucidated, some potential ones have been proposed: 1) Increased production of proinflammatory cytokines or chemokines can lead to immune-related damage in tissue which is anatomically prone. 2) Enhanced differentiation of lymphocytes containing T cells and B cells contributes to overpriming of T-cell-mediated immunity and overproduction of autoantibodies (Risbjerg et al., 2020; Ho et al., 2021). 3) Related to off-target effects of ICIs, hypophysitis induced by ipilimumab might be ascribed to targeting CTLA-4 expressed on pituitary tissues (Iwama et al., 2014). 4) The composition and percentage of the commensal microbiome may influence the curative effect for patients treated with ICI (Figure 2). The conclusion discovered from several kinds of research said that various irAEs were associated with the different superior microbiome, application of antibiotics was linked to poor prognosis, and fecal microbiota transplantation (FMT) could reduce immune colitis (Pierrard and Seront, 2019; Hommes et al., 2020; Andrews et al., 2021; Seton-Rogers, 2021). 5) Genetic susceptibility includes HLA haplotypes (Stamatouli et al., 2018).

FIGURE 2
www.frontiersin.org

FIGURE 2. Potential mechanisms of immune-related adverse events. 1) Blocking the interaction between PD-1 on T cells and PD-L1 on tumor cells may enhance the release of inflammatory cytokines from T cells. 2) Monoclonal antibodies, like anti-CTLA-4, may recognize antigen presented by the normal tissue (hypothalamic and pituitary tissues). 3) Overresponse of naive lymphocytes could proliferate autoreactive T cells and B cells. 4) The gut microbiome, which may be altered after ICI treatment, may influence T-cell function.

For the treatment of irAEs, there have been some guidelines providing algorithms for most of the frequently occurring irAEs. 1) Before ICI initiation, patients’ condition should be evaluated, including family history, general physical condition, and baseline laboratory tests (Ramos-Casals et al., 2020). 2) For those suffering grade I or II irAEs in hardly lethal organs, they could continue/hold immunotherapy. Otherwise, they would better take immunosuppressive or immune-modulating drugs, including corticosteroids, as first-line medicine to control irAEs and relieve clinical symptoms (Esfahani et al., 2020). 3) For those who may bring irreversible or fatal consequences, it is necessary to withhold ICIs and apply steroids or other immunosuppressants immediately (Brahmer et al., 2021). 4) Individual basis should be taken into account when resuming discontinued ICIs owing to irAEs. There are also artificial solutions such as developing engineering antibodies that can induce responsive immune defense and limit systemic exposure of CTLA-4 blockade at the same time (Pai et al., 2019; Lacouture et al., 2021).

Microbiome Related to ICI

With an estimated average of 3.8*1013 commensal bacterial resident in a 70 kg “reference man,” it is fluent in believing that gastrointestinal microbes play an important role in immunity (Sender et al., 2016). To date, there have been some oncogenic gut bacteria such as Salmonella typhi, Helicobacter spp., and Helicobacter pylori (Schwabe and Jobin, 2013; Gagnaire et al., 2017). On the contrary, some bacteria are thought to be beneficial for the proliferation of effector T cells and enhance antitumor efficacy (Pickard et al., 2017; Roy and Trinchieri, 2017). It is harder for mice supported in antibiotic exposed or germ-free conditions to benefit from CTLA-4 blockade versus those in specific pathogen-free environments (Vetizou et al., 2015). Thus, the linkage between microbiome and ICI needs to be elucidated (Table 2).

TABLE 2
www.frontiersin.org

TABLE 2. The role of immune checkpoints in bacteria-related diseases.

Clinical studies have reported that bacterial species can be differentially abundant in responders versus nonresponders (Katayama et al., 2019). Through feeding with B. fragilis, immunization with B. fragilis polysaccharides, or adoptive B. fragilis-specific T cells transfer, mice that failed in CTLA-4 blockade could regain their immunity. Transplantation of microbiota from melanoma patients to mice proved that B. fragilis favored the CTLA-4 blockade (Vetizou et al., 2015). In metastatic melanoma, Chaput et al. reported that patients with enriched Faecalibacterium and other Firmicutes as baseline microbiota presented a better prognosis than those with Bacteroides. However, the Bacteroidetes bring little colitis than Faecalibacterium (Chaput et al., 2017). In linkage with this, Gopalakrishnan et al. discovered that Faecalibacterium was enriched in responders, while Bacteroides thetaiotaomicron was enriched in nonresponders in melanoma patients (Gopalakrishnan et al., 2018). Using 16S ribosomal RNA gene sequencing, Matson et al. found out that Bifidobacterium longum, Collinsella aerofaciens, and Enterococcus faecium were more abundant in anti-PD-1 responders with metastatic melanoma (Matson et al., 2018).

The potential mechanisms through which the immune response is regulated by the microbiome may be as follows (Mazmanian et al., 2005; Helmink et al., 2019; Hayase and Jenq, 2021): 1) Through linkage between PAMPs and pattern recognized receptors (PRRs, such as Toll-like receptors), the adaptive immune response can be activated by APCs. 2) Cancer cells can bear cross-reactive neoantigens with microbiota, thus inducing an immune response. 3) Cytokines secreted by APCs or lymphocytes can be altered with specific metabolites or bacterial byproducts. 4) Metabolites entering the bloodstream could elicit a systemic response.

It also has been reported that irAEs induced by CTLA-4 occur most commonly and frequently at sites of the GI tract rich in bacteria. Disrupting the gut microbiota via antibiotics could potentially impair antitumor immune responses as well as response to immune checkpoint blockade (Helmink et al., 2019). Reconstruction of GI microbiome using FMT from healthy or responding donors shows a promising therapeutic effect with ICI-associated colitis relief and proportion of Tregs increase (Wang et al., 2018).

Still, the limitations of FMT should be taken into consideration. The connection between favorable microbiota and certain immune checkpoint blockade needs to be cleared. There could be adverse events induced by FMT, as we talked about above in IrAEs, either.

Immune Checkpoint Molecules in Virus-Infected Diseases

In chronic viral infection and cancer, due to long-term and low magnitude exposure to antigen, that T cell progressively loses its effector function with elevated coinhibitory receptor constitutive expression in order to diminish tissue damage is called “T-cell exhaustion.” Many pathogens and cancers promote inhibitory interactions to escape immune surveillance (Table 3). Thus, reversing the T-cell state is regarded as an effective solution in infectious diseases.

TABLE 3
www.frontiersin.org

TABLE 3. The role of immune checkpoints in virus infection diseases.

In mice with chronic LCMV infection, blockade of PD-1 restored CD8+ T cell function, suggesting that T-cell exhaustion is reversible. In patients with chronic hepatitis B, CTLA-4 blockade can reinvigorate hepatitis B virus- (HBV-) specific CD8+ T cells in both intrahepatic and peripheral compartments (Cao et al., 2018). With the coinhibition of PD-1 and CTLA-4, the effector function of HCV-specific CD8+ T cells can be restored in chronic hepatitis C patients (Cho et al., 2017). Meanwhile, inhibition of PD-1 can induce the production of cytokines (e.g., IFN-γ) in HIV/HBV-specific CD8+ T cells to enhance immune response (Jubel et al., 2020). Coexpressing with PD-1, LAG-3, TIM-3, and TIGIT blockade can also reverse dysfunctional T-cell responses and reduce cytokines production. It is widely known that TIM-3 is highly upregulated on virus and tumor Ag-specific CD8+ T cells, and antagonizing TIM-3 helps restore the function of CD8+ T cells (Clayton et al., 2014). Expression of LAG-3 has been reported to be associated with a reduction in invariant NKTs IFN-γ production during chronic HIV infection (Juno et al., 2015).

Discussion and Future Perspectives

Immune checkpoints are some vital regulators of the immune system. Now in most referred contexts, immune checkpoints are equivalent to inhibitor regulators of the immune system. Despite the immune checkpoint molecules that we have discussed above, there are still other immune checkpoint molecules, such as BTLA, KIR, A2AR, B7-H4, NOX2, HO-1, and SIGLEC7. Besides, the stimulatory immune checkpoints are also promising targets for immune therapy, such as CD40, CD122, CD137, OX40, and GITR. Relying on neoantigen expressed on tumor cells, T cells can target and exclude potential threats. So as to escape from host immunity, tumor cells requisition inhibitory molecules to bind and silence immune cells. The availability of immune checkpoint blockade as one of the effective supplemental methods for tumor treatment has been verified. However, some tumors show low immunogenicity and cannot respond effectively to immune checkpoint blockade. For initially responding tumors, selection of low immunogenic clones and inducement of tolerance due to tumor heterogeneity will develop frequent relapses and even hyperprogression in nonresponders, of which the range was between 4 and 29% (Denis et al., 2020). Such phenomenon is known as resistance (Sharma et al., 2017). The mechanisms of resistance can be divided into intrinsic and extrinsic (Figure 3). The intrinsic mechanisms are composed of lack of tumor antigen presentation, alteration of several inhibitory signaling pathways, and upregulation of other immune checkpoints. The extrinsic mechanisms are predominantly referred to as various elements in the TME (Baxter et al., 2021). To reverse the resistance and ameliorate patients’ symptoms, researchers came up with the idea to turn the “cold” immune response to “hot.” The strategies applied under such fundamental idea consist of turning down the volume of inhibitory immune signals, triggering T-cell priming, increasing the costimulatory signals, and modulation of the TME (Attili et al., 2021; Weiss and Sznol, 2021).

FIGURE 3
www.frontiersin.org

FIGURE 3. Mechanisms of resistance from ICI treatment. 1) β2M mutations lead to loss of HLA and antigen-presenting function. 2) Additional inhibitory signals expression. 3) Little tumor-infiltrating lymphocytes present in the tumor microenvironment resulting in nonresponse. 4) Immune suppressive cells in TME. 5) Loss of IFN-γ sensitivity. 6) Formation of low immunogenicity clone under selective pressure.

Meanwhile, the sailing of drug development is never smooth. Hundreds of clinical trials to develop new agents targeted at immune checkpoints have been terminated due to low responsiveness and fatal irAEs. IrAEs induced by ICI are an impassable mountain lying in front of us, with death as the most severe consequence. The clinical trial testing sym022 (anti-LAG-3 mAb) in humans with metastatic cancer, solid tumors, or lymphoma exhibits an unwanted outcome with high progression and irAEs rate (NCT03489369). In addition, the mechanisms under ICI still need to be shed light on.

In conclusion, despite the shortcomings of immune checkpoint blockade in clinical application, it is a promising strategy for cancer therapy, with a considerable proportion of applicants achieving an objective response. Further studies are needed to be explored to elucidate precise mechanisms, achieve potential will, and ameliorate adverse events to benefit more patients with tumors and other diseases.

Author Contributions

Literature search: HZ and YY, tables and figures: JY and YZ, writing the original manuscript: XC and YZ; editing manuscript: JY; review and editing manuscript: JL, MZ, and YZ.

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s Note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations or those of the publisher, the editors, and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.

References

Ahmadzadeh, M., Johnson, L. A., Heemskerk, B., Wunderlich, J. R., Dudley, M. E., White, D. E., et al. (2009). Tumor Antigen-specific CD8 T Cells Infiltrating the Tumor Express High Levels of PD-1 and Are Functionally Impaired. Blood 114 (8), 1537–1544. doi:10.1182/blood-2008-12-195792

PubMed Abstract | CrossRef Full Text | Google Scholar

Almutairi, A. R., McBride, A., Slack, M., Erstad, B. L., and Abraham, I. (2020). Potential Immune-Related Adverse Events Associated with Monotherapy and Combination Therapy of Ipilimumab, Nivolumab, and Pembrolizumab for Advanced Melanoma: A Systematic Review and Meta-Analysis. Front. Oncol. 10, 91. doi:10.3389/fonc.2020.00091

PubMed Abstract | CrossRef Full Text | Google Scholar

Andrews, M. C., Duong, C. P. M., Gopalakrishnan, V., Iebba, V., Chen, W.-S., Derosa, L., et al. (2021). Gut Microbiota Signatures Are Associated with Toxicity to Combined CTLA-4 and PD-1 Blockade. Nat. Med. 27 (8), 1432–1441. doi:10.1038/s41591-021-01406-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Ansell, S. M., Lesokhin, A. M., Borrello, I., Halwani, A., Scott, E. C., Gutierrez, M., et al. (2015). PD-1 Blockade with Nivolumab in Relapsed or Refractory Hodgkin's Lymphoma. N. Engl. J. Med. 372 (4), 311–319. doi:10.1056/NEJMoa1411087

CrossRef Full Text | Google Scholar

Ascierto, P. A., Del Vecchio, M., Robert, C., Mackiewicz, A., Chiarion-Sileni, V., Arance, A., et al. (2017). Ipilimumab 10 Mg/kg versus Ipilimumab 3 Mg/kg in Patients with Unresectable or Metastatic Melanoma: a Randomised, Double-Blind, Multicentre, Phase 3 Trial. Lancet Oncol. 18 (5), 611–622. doi:10.1016/S1470-2045(17)30231-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Attili, I., Tarantino, P., Passaro, A., Stati, V., Curigliano, G., and de Marinis, F. (2021). Strategies to Overcome Resistance to Immune Checkpoint Blockade in Lung Cancer. Lung Cancer 154, 151–160. doi:10.1016/j.lungcan.2021.02.035

PubMed Abstract | CrossRef Full Text | Google Scholar

Avery, L., Filderman, J., Szymczak-Workman, A. L., and Kane, L. P. (2018). Tim-3 Co-stimulation Promotes Short-Lived Effector T Cells, Restricts Memory Precursors, and Is Dispensable for T Cell Exhaustion. Proc. Natl. Acad. Sci. USA 115 (10), 2455–2460. doi:10.1073/pnas.1712107115

PubMed Abstract | CrossRef Full Text | Google Scholar

Baas, P., Scherpereel, A., Nowak, A. K., Fujimoto, N., Peters, S., Tsao, A. S., et al. (2021). First-line Nivolumab Plus Ipilimumab in Unresectable Malignant Pleural Mesothelioma (CheckMate 743): a Multicentre, Randomised, Open-Label, Phase 3 Trial. The Lancet 397 (10272), 375–386. doi:10.1016/S0140-6736(20)32714-8

CrossRef Full Text | Google Scholar

Baxter, M. A., Middleton, F., Cagney, H. P., and Petty, R. D. (2021). Resistance to Immune Checkpoint Inhibitors in Advanced Gastro-Oesophageal Cancers. Br. J. Cancer 125 (8), 1068–1079. doi:10.1038/s41416-021-01425-7

CrossRef Full Text | Google Scholar

Beckermann, K. E., Johnson, D. B., and Sosman, J. A. (2017). PD-1/PD-L1 Blockade in Renal Cell Cancer. Expert Rev. Clin. Immunol. 13 (1), 77–84. doi:10.1080/1744666X.2016.1214575

PubMed Abstract | CrossRef Full Text | Google Scholar

Berti, A., Bortolotti, R., Dipasquale, M., Kinspergher, S., Prokop, L., Grandi, G., et al. (2021). Meta-analysis of Immune-Related Adverse Events in Phase 3 Clinical Trials Assessing Immune Checkpoint Inhibitors for Lung Cancer. Crit. Rev. Oncology/Hematology 162, 103351. doi:10.1016/j.critrevonc.2021.103351

CrossRef Full Text | Google Scholar

Blackburn, S. D., Shin, H., Haining, W. N., Zou, T., Workman, C. J., Polley, A., et al. (2009). Coregulation of CD8+ T Cell Exhaustion by Multiple Inhibitory Receptors during Chronic Viral Infection. Nat. Immunol. 10 (1), 29–37. doi:10.1038/ni.1679

PubMed Abstract | CrossRef Full Text | Google Scholar

Brahmer, J. R., Abu-Sbeih, H., Ascierto, P. A., Brufsky, J., Cappelli, L. C., Cortazar, F. B., et al. (2021). Society for Immunotherapy of Cancer (SITC) Clinical Practice Guideline on Immune Checkpoint Inhibitor-Related Adverse Events. J. Immunother. Cancer 9 (6), e002435. doi:10.1136/jitc-2021-002435

CrossRef Full Text | Google Scholar

Brignone, C., Escudier, B., Grygar, C., Marcu, M., and Triebel, F. (2009). A Phase I Pharmacokinetic and Biological Correlative Study of IMP321, a Novel MHC Class II Agonist, in Patients with Advanced Renal Cell Carcinoma. Clin. Cancer Res. 15 (19), 6225–6231. doi:10.1158/1078-0432.CCR-09-0068

PubMed Abstract | CrossRef Full Text | Google Scholar

Cao, H., Zhang, R., and Zhang, W. (2018). CTLA-4 Interferes with the HBV-specific T cell Immune Response (Review). Int. J. Mol. Med. 42 (2), 703–712. doi:10.3892/ijmm.2018.3688

PubMed Abstract | CrossRef Full Text | Google Scholar

Casak, S. J., Marcus, L., Fashoyin-Aje, L., Mushti, S. L., Cheng, J., Shen, Y.-L., et al. (2021). FDA Approval Summary: Pembrolizumab for the First-Line Treatment of Patients with MSI-H/dMMR Advanced Unresectable or Metastatic Colorectal Carcinoma. Clin. Cancer Res. 27 (17), 4680–4684. doi:10.1158/1078-0432.CCR-21-0557

PubMed Abstract | CrossRef Full Text | Google Scholar

Chae, Y. K., Arya, A., Iams, W., Cruz, M. R., Chandra, S., Choi, J., et al. (2018). Current Landscape and Future of Dual Anti-CTLA4 and PD-1/pd-L1 Blockade Immunotherapy in Cancer; Lessons Learned from Clinical Trials with Melanoma and Non-small Cell Lung Cancer (NSCLC). J. Immunotherapy Cancer 6 (1), 39. doi:10.1186/s40425-018-0349-3

CrossRef Full Text | Google Scholar

Chaput, N., Lepage, P., Coutzac, C., Soularue, E., Le Roux, K., Monot, C., et al. (2017). Baseline Gut Microbiota Predicts Clinical Response and Colitis in Metastatic Melanoma Patients Treated with Ipilimumab. Ann. Oncol. 28 (6), 1368–1379. doi:10.1093/annonc/mdx108

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, D. S., and Mellman, I. (2017). Elements of Cancer Immunity and the Cancer-Immune Set point. Nature 541 (7637), 321–330. doi:10.1038/nature21349

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, Y., Zhou, Y., Tang, L., Peng, X., Jiang, H., Wang, G., et al. (2019). Immune-Checkpoint Inhibitors as the First Line Treatment of Advanced Non-small Cell Lung Cancer: A Meta-Analysis of Randomized Controlled Trials. J. Cancer 10 (25), 6261–6268. doi:10.7150/jca.34677

PubMed Abstract | CrossRef Full Text | Google Scholar

Chiba, S., Baghdadi, M., Akiba, H., Yoshiyama, H., Kinoshita, I., Dosaka-Akita, H., et al. (2012). Tumor-infiltrating DCs Suppress Nucleic Acid-Mediated Innate Immune Responses through Interactions between the Receptor TIM-3 and the Alarmin HMGB1. Nat. Immunol. 13 (9), 832–842. doi:10.1038/ni.2376

PubMed Abstract | CrossRef Full Text | Google Scholar

Chida, K., Kawazoe, A., Kawazu, M., Suzuki, T., Nakamura, Y., Nakatsura, T., et al. (2021). A Low Tumor Mutational Burden and PTEN Mutations Are Predictors of a Negative Response to PD-1 Blockade in MSI-H/dMMR Gastrointestinal Tumors. Clin. Cancer Res. 27 (13), 3714–3724. doi:10.1158/1078-0432.CCR-21-0401

PubMed Abstract | CrossRef Full Text | Google Scholar

Chihara, N., Madi, A., Kondo, T., Zhang, H., Acharya, N., Singer, M., et al. (2018). Induction and Transcriptional Regulation of the Co-inhibitory Gene Module in T Cells. Nature 558 (7710), 454–459. doi:10.1038/s41586-018-0206-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Cho, H., Kang, H., Lee, H., and Kim, C. (2017). Programmed Cell Death 1 (PD-1) and Cytotoxic T Lymphocyte-Associated Antigen 4 (CTLA-4) in Viral Hepatitis. Ijms 18 (7), 1517. doi:10.3390/ijms18071517

PubMed Abstract | CrossRef Full Text | Google Scholar

Choi, J., and Lee, S. Y. (2020). Clinical Characteristics and Treatment of Immune-Related Adverse Events of Immune Checkpoint Inhibitors. Immune Netw. 20 (1), e9. doi:10.4110/in.2020.20.e9

PubMed Abstract | CrossRef Full Text | Google Scholar

Clayton, K. L., Haaland, M. S., Douglas-Vail, M. B., Mujib, S., Chew, G. M., Ndhlovu, L. C., et al. (2014). T Cell Ig and Mucin Domain-Containing Protein 3 Is Recruited to the Immune Synapse, Disrupts Stable Synapse Formation, and Associates with Receptor Phosphatases. J.I. 192 (2), 782–791. doi:10.4049/jimmunol.1302663

CrossRef Full Text | Google Scholar

Connolly, C., Bambhania, K., and Naidoo, J. (2019). Immune-Related Adverse Events: A Case-Based Approach. Front. Oncol. 9, 530. doi:10.3389/fonc.2019.00530

PubMed Abstract | CrossRef Full Text | Google Scholar

Curran, M. A., Montalvo, W., Yagita, H., and Allison, J. P. (2010). PD-1 and CTLA-4 Combination Blockade Expands Infiltrating T Cells and Reduces Regulatory T and Myeloid Cells within B16 Melanoma Tumors. Proc. Natl. Acad. Sci. 107 (9), 4275–4280. doi:10.1073/pnas.0915174107

PubMed Abstract | CrossRef Full Text | Google Scholar

Darvin, P., Toor, S. M., Sasidharan Nair, V., and Elkord, E. (2018). Immune Checkpoint Inhibitors: Recent Progress and Potential Biomarkers. Exp. Mol. Med. 50 (12), 1–11. doi:10.1038/s12276-018-0191-1

CrossRef Full Text | Google Scholar

Davila, E., Kennedy, R., and Celis, E. (2003). Generation of Antitumor Immunity by Cytotoxic T Lymphocyte Epitope Peptide Vaccination, CpG-Oligodeoxynucleotide Adjuvant, and CTLA-4 Blockade. Cancer Res. 63 (12), 3281–3288.

PubMed Abstract | Google Scholar

Denis, M., Duruisseaux, M., Brevet, M., and Dumontet, C. (2020). How Can Immune Checkpoint Inhibitors Cause Hyperprogression in Solid Tumors. Front. Immunol. 11, 492. doi:10.3389/fimmu.2020.00492

PubMed Abstract | CrossRef Full Text | Google Scholar

Dermani, F. K., Samadi, P., Rahmani, G., Kohlan, A. K., and Najafi, R. (2019). PD‐1/PD‐L1 Immune Checkpoint: Potential Target for Cancer Therapy. J. Cel Physiol 234 (2), 1313–1325. doi:10.1002/jcp.27172

CrossRef Full Text | Google Scholar

Derré, L., Rivals, J.-P., Jandus, C., Pastor, S., Rimoldi, D., Romero, P., et al. (2010). BTLA Mediates Inhibition of Human Tumor-specific CD8+ T Cells that Can Be Partially Reversed by Vaccination. J. Clin. Invest. 120 (1), 157–167. doi:10.1172/JCI40070

CrossRef Full Text | Google Scholar

Dixon, K. O., Das, M., and Kuchroo, V. K. (2018). Human Disease Mutations Highlight the Inhibitory Function of TIM-3. Nat. Genet. 50 (12), 1640–1641. doi:10.1038/s41588-018-0289-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Doi, T., Ishikawa, T., Okayama, T., Oka, K., Mizushima, K., Yasuda, T., et al. (2017). The JAK/STAT Pathway Is Involved in the Upregulation of PD-L1 Expression in Pancreatic Cancer Cell Lines. Oncol. Rep. 37 (3), 1545–1554. doi:10.3892/or.2017.5399

PubMed Abstract | CrossRef Full Text | Google Scholar

Du, W., Yang, M., Turner, A., Xu, C., Ferris, R., Huang, J., et al. (2017). TIM-3 as a Target for Cancer Immunotherapy and Mechanisms of Action. Ijms 18 (3), 645. doi:10.3390/ijms18030645

PubMed Abstract | CrossRef Full Text | Google Scholar

Dyck, L., and Mills, K. H. G. (2017). Immune Checkpoints and Their Inhibition in Cancer and Infectious Diseases. Eur. J. Immunol. 47 (5), 765–779. doi:10.1002/eji.201646875

CrossRef Full Text | Google Scholar

Ebert, P. J. R., Cheung, J., Yang, Y., McNamara, E., Hong, R., Moskalenko, M., et al. (2016). MAP Kinase Inhibition Promotes T Cell and Anti-tumor Activity in Combination with PD-L1 Checkpoint Blockade. Immunity 44 (3), 609–621. doi:10.1016/j.immuni.2016.01.024

PubMed Abstract | CrossRef Full Text | Google Scholar

Emens, L. A., Adams, S., Barrios, C. H., Diéras, V., Iwata, H., Loi, S., et al. (2021). First-line Atezolizumab Plus Nab-Paclitaxel for Unresectable, Locally Advanced, or Metastatic Triple-Negative Breast Cancer: IMpassion130 Final Overall Survival Analysis. Ann. Oncol. 32 (8), 983–993. doi:10.1016/j.annonc.2021.05.355

PubMed Abstract | CrossRef Full Text | Google Scholar

Esfahani, K., Elkrief, A., Calabrese, C., Lapointe, R., Hudson, M., Routy, B., et al. (2020). Moving towards Personalized Treatments of Immune-Related Adverse Events. Nat. Rev. Clin. Oncol. 17 (8), 504–515. doi:10.1038/s41571-020-0352-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Finkelmeier, F., Waidmann, O., and Trojan, J. (2018). Nivolumab for the Treatment of Hepatocellular Carcinoma. Expert Rev. Anticancer Ther. 18 (12), 1169–1175. doi:10.1080/14737140.2018.1535315

PubMed Abstract | CrossRef Full Text | Google Scholar

Flies, D. B., Han, X., Higuchi, T., Zheng, L., Sun, J., Ye, J. J., et al. (2014). Coinhibitory Receptor PD-1H Preferentially Suppresses CD4+ T Cell-Mediated Immunity. J. Clin. Invest. 124 (5), 1966–1975. doi:10.1172/JCI74589

CrossRef Full Text | Google Scholar

Freeman, G. J., Long, A. J., Iwai, Y., Bourque, K., Chernova, T., Nishimura, H., et al. (2000). Engagement of the PD-1 Immunoinhibitory Receptor by a Novel B7 Family Member Leads to Negative Regulation of Lymphocyte Activation. J. Exp. Med. 192 (7), 1027–1034. doi:10.1084/jem.192.7.1027

CrossRef Full Text | Google Scholar

Gagnaire, A., Nadel, B., Raoult, D., Neefjes, J., and Gorvel, J.-P. (2017). Collateral Damage: Insights into Bacterial Mechanisms that Predispose Host Cells to Cancer. Nat. Rev. Microbiol. 15 (2), 109–128. doi:10.1038/nrmicro.2016.171

PubMed Abstract | CrossRef Full Text | Google Scholar

Gleason, M. K., Lenvik, T. R., McCullar, V., Felices, M., O'Brien, M. S., Cooley, S. A., et al. (2012). Tim-3 Is an Inducible Human Natural Killer Cell Receptor that Enhances Interferon Gamma Production in Response to Galectin-9. Blood 119 (13), 3064–3072. doi:10.1182/blood-2011-06-360321

PubMed Abstract | CrossRef Full Text | Google Scholar

Goldberg, M. V., and Drake, C. G. (2010). LAG-3 in Cancer Immunotherapy. Curr. Top. Microbiol. Immunol. 344, 269–278. doi:10.1007/82_2010_114

PubMed Abstract | CrossRef Full Text | Google Scholar

Gopalakrishnan, V., Spencer, C. N., Nezi, L., Reuben, A., Andrews, M. C., Karpinets, T. V., et al. (2018). Gut Microbiome Modulates Response to Anti-PD-1 Immunotherapy in Melanoma Patients. Science 359 (6371), 97–103. doi:10.1126/science.aan4236

PubMed Abstract | CrossRef Full Text | Google Scholar

Han, X., Vesely, M. D., Yang, W., Sanmamed, M. F., Badri, T., Alawa, J., et al. (2019). PD-1H (VISTA)-mediated Suppression of Autoimmunity in Systemic and Cutaneous Lupus Erythematosus. Sci. Transl. Med. 11 (522), eaax1159. doi:10.1126/scitranslmed.aax1159

PubMed Abstract | CrossRef Full Text | Google Scholar

Han, Y., Liu, D., and Li, L. (2020). PD-1/PD-L1 Pathway: Current Researches in Cancer. Am. J. Cancer Res. 10 (3), 727–742.

Google Scholar

Hayase, E., and Jenq, R. R. (2021). Role of the Intestinal Microbiome and Microbial-Derived Metabolites in Immune Checkpoint Blockade Immunotherapy of Cancer. Genome Med. 13 (1), 107. doi:10.1186/s13073-021-00923-w

PubMed Abstract | CrossRef Full Text | Google Scholar

Helmink, B. A., Khan, M. A. W., Hermann, A., Gopalakrishnan, V., and Wargo, J. A. (2019). The Microbiome, Cancer, and Cancer Therapy. Nat. Med. 25 (3), 377–388. doi:10.1038/s41591-019-0377-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Henry, K. E., Mack, K. N., Nagle, V. L., Cornejo, M., Michel, A. O., Fox, I. L., et al. (2021). ERK Inhibition Improves Anti-PD-L1 Immune Checkpoint Blockade in Preclinical Pancreatic Ductal Adenocarcinoma. Mol. Cancer Ther. 20, 2026–2034. doi:10.1158/1535-7163.MCT-20-1112

PubMed Abstract | CrossRef Full Text | Google Scholar

Ho, A. K., Ho, A. M.-H., Cooksley, T., Nguyen, G., Erb, J., and Mizubuti, G. B. (2021). Immune-Related Adverse Events Associated with Immune Checkpoint Inhibitor Therapy. Anesth. Analg 132 (2), 374–383. doi:10.1213/ANE.0000000000005029

PubMed Abstract | CrossRef Full Text | Google Scholar

Hodi, F. S., O'Day, S. J., McDermott, D. F., Weber, R. W., Sosman, J. A., Haanen, J. B., et al. (2010). Improved Survival with Ipilimumab in Patients with Metastatic Melanoma. N. Engl. J. Med. 363 (8), 711–723. doi:10.1056/NEJMoa1003466

CrossRef Full Text | Google Scholar

Hommes, J. W., Verheijden, R. J., Suijkerbuijk, K. P. M., and Hamann, D. (2020). Biomarkers of Checkpoint Inhibitor Induced Immune-Related Adverse Events-A Comprehensive Review. Front. Oncol. 10, 585311. doi:10.3389/fonc.2020.585311

PubMed Abstract | CrossRef Full Text | Google Scholar

Hosseinkhani, N., Derakhshani, A., Shadbad, M. A., Argentiero, A., Racanelli, V., Kazemi, T., et al. (2021). The Role of V-Domain Ig Suppressor of T Cell Activation (VISTA) in Cancer Therapy: Lessons Learned and the Road Ahead. Front. Immunol. 12, 676181. doi:10.3389/fimmu.2021.676181

PubMed Abstract | CrossRef Full Text | Google Scholar

Hotchkiss, R. S., and Moldawer, L. L. (2014). Parallels between Cancer and Infectious Disease. N. Engl. J. Med. 371 (4), 380–383. doi:10.1056/NEJMcibr1404664

CrossRef Full Text | Google Scholar

Hsu, J., Hodgins, J. J., Marathe, M., Nicolai, C. J., Bourgeois-Daigneault, M.-C., Trevino, T. N., et al. (2018). Contribution of NK Cells to Immunotherapy Mediated by PD-1/pd-L1 Blockade. J. Clin. Invest. 128 (10), 4654–4668. doi:10.1172/JCI99317

CrossRef Full Text | Google Scholar

Huang, C.-T., Workman, C. J., Flies, D., Pan, X., Marson, A. L., Zhou, G., et al. (2004). Role of LAG-3 in Regulatory T Cells. Immunity 21 (4), 503–513. doi:10.1016/j.immuni.2004.08.010

PubMed Abstract | CrossRef Full Text | Google Scholar

Huang, X., Zhang, X., Li, E., Zhang, G., Wang, X., Tang, T., et al. (2020). VISTA: an Immune Regulatory Protein Checking Tumor and Immune Cells in Cancer Immunotherapy. J. Hematol. Oncol. 13 (1), 83. doi:10.1186/s13045-020-00917-y

CrossRef Full Text | Google Scholar

Huang, Y.-H., Zhu, C., Kondo, Y., Anderson, A. C., Gandhi, A., Russell, A., et al. (2015). CEACAM1 Regulates TIM-3-Mediated Tolerance and Exhaustion. Nature 517 (7534), 386–390. doi:10.1038/nature13848

PubMed Abstract | CrossRef Full Text | Google Scholar

Iwama, S., De Remigis, A., Callahan, M. K., Slovin, S. F., Wolchok, J. D., and Caturegli, P. (2014). Pituitary Expression of CTLA-4 Mediates Hypophysitis Secondary to Administration of CTLA-4 Blocking Antibody. Sci. Transl. Med. 6 (230), 230ra245. doi:10.1126/scitranslmed.3008002

CrossRef Full Text | Google Scholar

Janjigian, Y. Y., Shitara, K., Moehler, M., Garrido, M., Salman, P., Shen, L., et al. (2021). First-line Nivolumab Plus Chemotherapy versus Chemotherapy Alone for Advanced Gastric, Gastro-Oesophageal junction, and Oesophageal Adenocarcinoma (CheckMate 649): a Randomised, Open-Label, Phase 3 Trial. Lancet 398 (10294), 27–40. doi:10.1016/S0140-6736(21)00797-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Johnston, R. J., Su, L. J., Pinckney, J., Critton, D., Boyer, E., Krishnakumar, A., et al. (2019). VISTA Is an Acidic pH-Selective Ligand for PSGL-1. Nature 574 (7779), 565–570. doi:10.1038/s41586-019-1674-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Jubel, J. M., Barbati, Z. R., Burger, C., Wirtz, D. C., and Schildberg, F. A. (2020). The Role of PD-1 in Acute and Chronic Infection. Front. Immunol. 11, 487. doi:10.3389/fimmu.2020.00487

PubMed Abstract | CrossRef Full Text | Google Scholar

Juno, J. A., Stalker, A. T., Waruk, J. L., Oyugi, J., Kimani, M., Plummer, F. A., et al. (2015). Elevated Expression of LAG-3, but Not PD-1, Is Associated with Impaired iNKT Cytokine Production during Chronic HIV-1 Infection and Treatment. Retrovirology 12, 17. doi:10.1186/s12977-015-0142-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Katayama, Y., Yamada, T., Shimamoto, T., Iwasaku, M., Kaneko, Y., Uchino, J., et al. (2019). The Role of the Gut Microbiome on the Efficacy of Immune Checkpoint Inhibitors in Japanese Responder Patients with Advanced Non-small Cell Lung Cancer. Transl Lung Cancer Res. 8 (6), 847–853. doi:10.21037/tlcr.2019.10.23

PubMed Abstract | CrossRef Full Text | Google Scholar

Khan, M., Arooj, S., and Wang, H. (2020). NK Cell-Based Immune Checkpoint Inhibition. Front. Immunol. 11, 167. doi:10.3389/fimmu.2020.00167

PubMed Abstract | CrossRef Full Text | Google Scholar

Kikushige, Y., Miyamoto, T., Yuda, J., Jabbarzadeh-Tabrizi, S., Shima, T., Takayanagi, S.-i., et al. (2015). A TIM-3/Gal-9 Autocrine Stimulatory Loop Drives Self-Renewal of Human Myeloid Leukemia Stem Cells and Leukemic Progression. Cell Stem Cell 17 (3), 341–352. doi:10.1016/j.stem.2015.07.011

PubMed Abstract | CrossRef Full Text | Google Scholar

Kim, K. J., Lee, H. W., and Seong, J. (2021). Combination Therapy with anti‐T‐cell Immunoglobulin and Mucin‐domain Containing Molecule 3 and Radiation Improves Antitumor Efficacy in Murine Hepatocellular Carcinoma. J. Gastroenterol. Hepatol. 36 (5), 1357–1365. doi:10.1111/jgh.15319

CrossRef Full Text | Google Scholar

Kong, K.-F., Fu, G., Zhang, Y., Yokosuka, T., Casas, J., Canonigo-Balancio, A. J., et al. (2014). Protein Kinase C-η Controls CTLA-4-Mediated Regulatory T Cell Function. Nat. Immunol. 15 (5), 465–472. doi:10.1038/ni.2866

PubMed Abstract | CrossRef Full Text | Google Scholar

Kouo, T., Huang, L., Pucsek, A. B., Cao, M., Solt, S., Armstrong, T., et al. (2015). Galectin-3 Shapes Antitumor Immune Responses by Suppressing CD8+ T Cells via LAG-3 and Inhibiting Expansion of Plasmacytoid Dendritic Cells. Cancer Immunol. Res. 3 (4), 412–423. doi:10.1158/2326-6066.CIR-14-0150

PubMed Abstract | CrossRef Full Text | Google Scholar

Lacouture, M. E., Sibaud, V., Gerber, P. A., van den Hurk, C., Fernández-Peñas, P., Santini, D., et al. (2021). Prevention and Management of Dermatological Toxicities Related to Anticancer Agents: ESMO Clinical Practice Guidelines☆. Ann. Oncol. 32 (2), 157–170. doi:10.1016/j.annonc.2020.11.005

PubMed Abstract | CrossRef Full Text | Google Scholar

Larkin, J., Chiarion-Sileni, V., Gonzalez, R., Grob, J. J., Cowey, C. L., Lao, C. D., et al. (2015). Combined Nivolumab and Ipilimumab or Monotherapy in Untreated Melanoma. N. Engl. J. Med. 373 (1), 23–34. doi:10.1056/NEJMoa1504030

PubMed Abstract | CrossRef Full Text | Google Scholar

Leach, D. R., Krummel, M. F., and Allison, J. P. (1996). Enhancement of Antitumor Immunity by CTLA-4 Blockade. Science 271 (5256), 1734–1736. doi:10.1126/science.271.5256.1734

PubMed Abstract | CrossRef Full Text | Google Scholar

Lecocq, Q., Keyaerts, M., Devoogdt, N., and Breckpot, K. (2020). The Next-Generation Immune Checkpoint LAG-3 and its Therapeutic Potential in Oncology: Third Time's a Charm. Ijms 22 (1), 75. doi:10.3390/ijms22010075

CrossRef Full Text | Google Scholar

Lenschow, D. J., and Bluestone, J. A. (1993). T Cell Co-stimulation and In Vivo Tolerance. Curr. Opin. Immunol. 5 (5), 747–752. doi:10.1016/0952-7915(93)90132-c

PubMed Abstract | CrossRef Full Text | Google Scholar

Lenschow, D. J., Walunas, T. L., and Bluestone, J. A. (1996). CD28/B7 System of T Cell Costimulation. Annu. Rev. Immunol. 14, 233–258. doi:10.1146/annurev.immunol.14.1.233

PubMed Abstract | CrossRef Full Text | Google Scholar

Lin, W., Chen, M., Hong, L., Zhao, H., and Chen, Q. (2018). Crosstalk between PD-1/pd-L1 Blockade and its Combinatorial Therapies in Tumor Immune Microenvironment: A Focus on HNSCC. Front. Oncol. 8, 532. doi:10.3389/fonc.2018.00532

PubMed Abstract | CrossRef Full Text | Google Scholar

Lines, J. L., Pantazi, E., Mak, J., Sempere, L. F., Wang, L., O'Connell, S., et al. (2014). VISTA Is an Immune Checkpoint Molecule for Human T Cells. Cancer Res. 74 (7), 1924–1932. doi:10.1158/0008-5472.CAN-13-1504

PubMed Abstract | CrossRef Full Text | Google Scholar

Linsley, P. S., Brady, W., Urnes, M., Grosmaire, L. S., Damle, N. K., and Ledbetter, J. A. (1991). CTLA-4 Is a Second Receptor for the B Cell Activation Antigen B7. J. Exp. Med. 174 (3), 561–569. doi:10.1084/jem.174.3.561

CrossRef Full Text | Google Scholar

Linsley, P. S., Greene, J. L., Brady, W., Bajorath, J., Ledbetter, J. A., and Peach, R. (1994). Human B7-1 (CD80) and B7-2 (CD86) Bind with Similar Avidities but Distinct Kinetics to CD28 and CTLA-4 Receptors. Immunity 1 (9), 793–801. doi:10.1016/s1074-7613(94)80021-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Lipson, E. J., Tawbi, H. A.-H., Schadendorf, D., Ascierto, P. A., Matamala, L., Gutiérrez, E. C., et al. (2021). Relatlimab (RELA) Plus Nivolumab (NIVO) Versus NIVO in First-Line Advanced Melanoma: Primary Phase III Results from RELATIVITY-047 (CA224-047). J. Clin. Oncol. 39, 9503. doi:10.1200/JCO.2021.39.15_suppl.9503

CrossRef Full Text | Google Scholar

Liu, J.-F., Wu, L., Yang, L.-L., Deng, W.-W., Mao, L., Wu, H., et al. (2018). Blockade of TIM3 Relieves Immunosuppression through Reducing Regulatory T Cells in Head and Neck Cancer. J. Exp. Clin. Cancer Res. 37 (1), 44. doi:10.1186/s13046-018-0713-7

CrossRef Full Text | Google Scholar

Liu, J., Yuan, Y., Chen, W., Putra, J., Suriawinata, A. A., Schenk, A. D., et al. (2015). Immune-checkpoint Proteins VISTA and PD-1 Nonredundantly Regulate Murine T-Cell Responses. Proc. Natl. Acad. Sci. USA 112 (21), 6682–6687. doi:10.1073/pnas.1420370112

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, M., Wei, F., Wang, J., Yu, W., Shen, M., Liu, T., et al. (2021). Myeloid-derived Suppressor Cells Regulate the Immunosuppressive Functions of PD-1−pd-L1+ Bregs through PD-L1/PI3K/AKT/NF-κB axis in Breast Cancer. Cell Death Dis 12 (5), 465. doi:10.1038/s41419-021-03745-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Lo, B., Zhang, K., Lu, W., Zheng, L., Zhang, Q., Kanellopoulou, C., et al. (2015). Patients with LRBA Deficiency Show CTLA4 Loss and Immune Dysregulation Responsive to Abatacept Therapy. Science 349 (6246), 436–440. doi:10.1126/science.aaa1663

PubMed Abstract | CrossRef Full Text | Google Scholar

Mao, X., Ou, M. T., Karuppagounder, S. S., Kam, T.-I., Yin, X., Xiong, Y., et al. (2016). Pathological α-synuclein Transmission Initiated by Binding Lymphocyte-Activation Gene 3. Science 353 (6307), aah3374. doi:10.1126/science.aah3374

PubMed Abstract | CrossRef Full Text | Google Scholar

Maruhashi, T., Sugiura, D., Okazaki, I.-m., and Okazaki, T. (2020). LAG-3: from Molecular Functions to Clinical Applications. J. Immunother. Cancer 8 (2), e001014. doi:10.1136/jitc-2020-001014

CrossRef Full Text | Google Scholar

Matson, V., Fessler, J., Bao, R., Chongsuwat, T., Zha, Y., Alegre, M.-L., et al. (2018). The Commensal Microbiome Is Associated with Anti-PD-1 Efficacy in Metastatic Melanoma Patients. Science 359 (6371), 104–108. doi:10.1126/science.aao3290

PubMed Abstract | CrossRef Full Text | Google Scholar

Mazmanian, S. K., Liu, C. H., Tzianabos, A. O., and Kasper, D. L. (2005). An Immunomodulatory Molecule of Symbiotic Bacteria Directs Maturation of the Host Immune System. Cell 122 (1), 107–118. doi:10.1016/j.cell.2005.05.007

PubMed Abstract | CrossRef Full Text | Google Scholar

McKay, R. R., McGregor, B. A., Xie, W., Braun, D. A., Wei, X., Kyriakopoulos, C. E., et al. (2020). Optimized Management of Nivolumab and Ipilimumab in Advanced Renal Cell Carcinoma: A Response-Based Phase II Study (OMNIVORE). Jco 38 (36), 4240–4248. doi:10.1200/JCO.20.02295

PubMed Abstract | CrossRef Full Text | Google Scholar

Metz, R., Rust, S., Duhadaway, J. B., Mautino, M. R., Munn, D. H., Vahanian, N. N., et al. (2012). Ido Inhibits a Tryptophan Sufficiency Signal that Stimulates mTOR: A Novel Ido Effector Pathway Targeted by D-1-Methyl-Tryptophan. Oncoimmunology 1 (9), 1460–1468. doi:10.4161/onci.21716

PubMed Abstract | CrossRef Full Text | Google Scholar

Mezrich, J. D., Fechner, J. H., Zhang, X., Johnson, B. P., Burlingham, W. J., and Bradfield, C. A. (2010). An Interaction between Kynurenine and the Aryl Hydrocarbon Receptor Can Generate Regulatory T Cells. J.I. 185 (6), 3190–3198. doi:10.4049/jimmunol.0903670

CrossRef Full Text | Google Scholar

Monney, L., Sabatos, C. A., Gaglia, J. L., Ryu, A., Waldner, H., Chernova, T., et al. (2002). Th1-specific Cell Surface Protein Tim-3 Regulates Macrophage Activation and Severity of an Autoimmune Disease. Nature 415 (6871), 536–541. doi:10.1038/415536a

PubMed Abstract | CrossRef Full Text | Google Scholar

Muller, S., Victoria Lai, W., Adusumilli, P. S., Desmeules, P., Frosina, D., Jungbluth, A., et al. (2020). V-domain Ig-Containing Suppressor of T-Cell Activation (VISTA), a Potentially Targetable Immune Checkpoint Molecule, Is Highly Expressed in Epithelioid Malignant Pleural Mesothelioma. Mod. Pathol. 33 (2), 303–311. doi:10.1038/s41379-019-0364-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Munn, D. H., and Mellor, A. L. (2013). Indoleamine 2,3 Dioxygenase and Metabolic Control of Immune Responses. Trends Immunol. 34 (3), 137–143. doi:10.1016/j.it.2012.10.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Munn, D. H., Sharma, M. D., Baban, B., Harding, H. P., Zhang, Y., Ron, D., et al. (2005). GCN2 Kinase in T Cells Mediates Proliferative Arrest and Anergy Induction in Response to Indoleamine 2,3-dioxygenase. Immunity 22 (5), 633–642. doi:10.1016/j.immuni.2005.03.013

PubMed Abstract | CrossRef Full Text | Google Scholar

Murphy, K. M., Nelson, C. A., and Šedý, J. R. (2006). Balancing Co-stimulation and Inhibition with BTLA and HVEM. Nat. Rev. Immunol. 6 (9), 671–681. doi:10.1038/nri1917

PubMed Abstract | CrossRef Full Text | Google Scholar

Nakayama, M., Akiba, H., Takeda, K., Kojima, Y., Hashiguchi, M., Azuma, M., et al. (2009). Tim-3 Mediates Phagocytosis of Apoptotic Cells and Cross-Presentation. Blood 113 (16), 3821–3830. doi:10.1182/blood-2008-10-185884

PubMed Abstract | CrossRef Full Text | Google Scholar

Nandi, D., Pathak, S., Verma, T., Singh, M., Chattopadhyay, A., Thakur, S., et al. (2020). T Cell Costimulation, Checkpoint Inhibitors and Anti-tumor Therapy. J. Biosci. 45, 50. doi:10.1007/s12038-020-0020-2

CrossRef Full Text | Google Scholar

Nassar, A. H., Mouw, K. W., Jegede, O., Shinagare, A. B., Kim, J., Liu, C.-J., et al. (2020). A Model Combining Clinical and Genomic Factors to Predict Response to PD-1/pd-L1 Blockade in Advanced Urothelial Carcinoma. Br. J. Cancer 122 (4), 555–563. doi:10.1038/s41416-019-0686-0

CrossRef Full Text | Google Scholar

Neel, B. G., Gu, H., and Pao, L. (2003). The 'Shp'ing News: SH2 Domain-Containing Tyrosine Phosphatases in Cell Signaling. Trends Biochem. Sci. 28 (6), 284–293. doi:10.1016/S0968-0004(03)00091-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Nogueira-Machado, J. A., Volpe, C. M. d. O., Veloso, C. A., and Chaves, M. M. (2011). HMGB1, TLR and RAGE: a Functional Tripod that Leads to Diabetic Inflammation. Expert Opin. Ther. Targets 15 (8), 1023–1035. doi:10.1517/14728222.2011.575360

PubMed Abstract | CrossRef Full Text | Google Scholar

Oliveira, A. F., Bretes, L., and Furtado, I. (2019). Review of PD-1/pd-L1 Inhibitors in Metastatic dMMR/MSI-H Colorectal Cancer. Front. Oncol. 9, 396. doi:10.3389/fonc.2019.00396

PubMed Abstract | CrossRef Full Text | Google Scholar

Olsson, C., Riebeck, K., Dohlsten, M., and Michaëlsson, E. (1999). CTLA-4 Ligation Suppresses CD28-Induced NF-Κb and AP-1 Activity in Mouse T Cell Blasts. J. Biol. Chem. 274 (20), 14400–14405. doi:10.1074/jbc.274.20.14400

PubMed Abstract | CrossRef Full Text | Google Scholar

Pagès, F., Ragueneau, M., Rottapel, R., Truneh, A., Nunes, J., Imbert, J., et al. (1994). Binding of Phosphatidyl-Inositol-3-OH Kinase to CD28 Is Required for T-Cell Signalling. Nature 369 (6478), 327–329. doi:10.1038/369327a0

PubMed Abstract | CrossRef Full Text | Google Scholar

Pai, C.-C. S., Simons, D. M., Lu, X., Evans, M., Wei, J., Wang, Y.-h., et al. (2018). Tumor-conditional Anti-CTLA4 Uncouples Antitumor Efficacy from Immunotherapy-Related Toxicity. J. Clin. Invest. 129 (1), 349–363. doi:10.1172/JCI123391

CrossRef Full Text | Google Scholar

Panjwani, P. K., Charu, V., DeLisser, M., Molina-Kirsch, H., Natkunam, Y., and Zhao, S. (2018). Programmed Death-1 Ligands PD-L1 and PD-L2 Show Distinctive and Restricted Patterns of Expression in Lymphoma Subtypes. Hum. Pathol. 71, 91–99. doi:10.1016/j.humpath.2017.10.029

PubMed Abstract | CrossRef Full Text | Google Scholar

Patel, R., Bock, M., Polotti, C. F., and Elsamra, S. (2017). Pharmacokinetic Drug Evaluation of Atezolizumab for the Treatment of Locally Advanced or Metastatic Urothelial Carcinoma. Expert Opin. Drug Metab. Toxicol. 13 (2), 225–232. doi:10.1080/17425255.2017.1277204

PubMed Abstract | CrossRef Full Text | Google Scholar

Patsoukis, N., Duke-Cohan, J. S., Chaudhri, A., Aksoylar, H.-I., Wang, Q., Council, A., et al. (2020). Interaction of SHP-2 SH2 Domains with PD-1 ITSM Induces PD-1 Dimerization and SHP-2 Activation. Commun. Biol. 3 (1), 128. doi:10.1038/s42003-020-0845-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Pauken, K. E., and Wherry, E. J. (2015). Overcoming T Cell Exhaustion in Infection and Cancer. Trends Immunol. 36 (4), 265–276. doi:10.1016/j.it.2015.02.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Peggs, K. S., Quezada, S. A., Korman, A. J., and Allison, J. P. (2006). Principles and Use of Anti-CTLA4 Antibody in Human Cancer Immunotherapy. Curr. Opin. Immunol. 18 (2), 206–213. doi:10.1016/j.coi.2006.01.011

PubMed Abstract | CrossRef Full Text | Google Scholar

Pickard, J. M., Zeng, M. Y., Caruso, R., and Núñez, G. (2017). Gut Microbiota: Role in Pathogen Colonization, Immune Responses, and Inflammatory Disease. Immunol. Rev. 279 (1), 70–89. doi:10.1111/imr.12567

PubMed Abstract | CrossRef Full Text | Google Scholar

Pierrard, J., and Seront, E. (2019). Impact of the Gut Microbiome on Immune Checkpoint Inhibitor Efficacy-A Systematic Review. Curr. Oncol. 26 (6), 395–403. doi:10.3747/co.26.5177

PubMed Abstract | CrossRef Full Text | Google Scholar

Pinto, J. A., Raez, L. E., Oliveres, H., and Rolfo, C. C. (2019). Current Knowledge of Ipilimumab and its Use in Treating Non-small Cell Lung Cancer. Expert Opin. Biol. Ther. 19 (6), 509–515. doi:10.1080/14712598.2019.1610380

PubMed Abstract | CrossRef Full Text | Google Scholar

Prasad, V., and Kaestner, V. (2017). Nivolumab and Pembrolizumab: Monoclonal Antibodies against Programmed Cell Death-1 (PD-1) that Are Interchangeable. Semin. Oncol. 44 (2), 132–135. doi:10.1053/j.seminoncol.2017.06.007

PubMed Abstract | CrossRef Full Text | Google Scholar

Qureshi, O. S., Kaur, S., Hou, T. Z., Jeffery, L. E., Poulter, N. S., Briggs, Z., et al. (2012). Constitutive Clathrin-Mediated Endocytosis of CTLA-4 Persists during T Cell Activation. J. Biol. Chem. 287 (12), 9429–9440. doi:10.1074/jbc.M111.304329

CrossRef Full Text | Google Scholar

Qureshi, O. S., Zheng, Y., Nakamura, K., Attridge, K., Manzotti, C., Schmidt, E. M., et al. (2011). Trans-endocytosis of CD80 and CD86: a Molecular Basis for the Cell-Extrinsic Function of CTLA-4. Science 332 (6029), 600–603. doi:10.1126/science.1202947

PubMed Abstract | CrossRef Full Text | Google Scholar

Ramos-Casals, M., Brahmer, J. R., Callahan, M. K., Flores-Chávez, A., Keegan, N., Khamashta, M. A., et al. (2020). Immune-related Adverse Events of Checkpoint Inhibitors. Nat. Rev. Dis. Primers 6 (1), 38. doi:10.1038/s41572-020-0160-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Rangachari, M., Zhu, C., Sakuishi, K., Xiao, S., Karman, J., Chen, A., et al. (2012). Bat3 Promotes T Cell Responses and Autoimmunity by Repressing Tim-3-Mediated Cell Death and Exhaustion. Nat. Med. 18 (9), 1394–1400. doi:10.1038/nm.2871

PubMed Abstract | CrossRef Full Text | Google Scholar

Remon, J., and Besse, B. (2017). Immune Checkpoint Inhibitors in First-Line Therapy of Advanced Non-small Cell Lung Cancer. Curr. Opin. Oncol. 29 (2), 97–104. doi:10.1097/CCO.0000000000000351

PubMed Abstract | CrossRef Full Text | Google Scholar

Risbjerg, R. S., Hansen, M. V., Sørensen, A. S., and Kragstrup, T. W. (2020). The Effects of B Cell Depletion on Immune Related Adverse Events Associated with Immune Checkpoint Inhibition. Exp. Hematol. Oncol. 9, 9. doi:10.1186/s40164-020-00167-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Ritprajak, P., and Azuma, M. (2015). Intrinsic and Extrinsic Control of Expression of the Immunoregulatory Molecule PD-L1 in Epithelial Cells and Squamous Cell Carcinoma. Oral Oncol. 51 (3), 221–228. doi:10.1016/j.oraloncology.2014.11.014

PubMed Abstract | CrossRef Full Text | Google Scholar

Romano, E., Kusio-Kobialka, M., Foukas, P. G., Baumgaertner, P., Meyer, C., Ballabeni, P., et al. (2015). Ipilimumab-dependent Cell-Mediated Cytotoxicity of Regulatory T Cells Ex Vivo by Nonclassical Monocytes in Melanoma Patients. Proc. Natl. Acad. Sci. USA 112 (19), 6140–6145. doi:10.1073/pnas.1417320112

PubMed Abstract | CrossRef Full Text | Google Scholar

Rowshanravan, B., Halliday, N., and Sansom, D. M. (2018). CTLA-4: a Moving Target in Immunotherapy. Blood 131 (1), 58–67. doi:10.1182/blood-2017-06-741033

PubMed Abstract | CrossRef Full Text | Google Scholar

Roy, S., and Trinchieri, G. (2017). Microbiota: a Key Orchestrator of Cancer Therapy. Nat. Rev. Cancer 17 (5), 271–285. doi:10.1038/nrc.2017.13

PubMed Abstract | CrossRef Full Text | Google Scholar

Ruffo, E., Wu, R. C., Bruno, T. C., Workman, C. J., and Vignali, D. A. A. (2019). Lymphocyte-activation Gene 3 (LAG3): The Next Immune Checkpoint Receptor. Semin. Immunol. 42, 101305. doi:10.1016/j.smim.2019.101305

PubMed Abstract | CrossRef Full Text | Google Scholar

Saito, H., Kono, Y., Murakami, Y., Shishido, Y., Kuroda, H., Matsunaga, T., et al. (2018). Highly Activated PD-1/pd-L1 Pathway in Gastric Cancer with PD-L1 Expression. Ar 38 (1), 107–112. doi:10.21873/anticanres.12197

PubMed Abstract | CrossRef Full Text | Google Scholar

Sandigursky, S., and Mor, A. (2018). Immune-Related Adverse Events in Cancer Patients Treated with Immune Checkpoint Inhibitors. Curr. Rheumatol. Rep. 20 (10), 65. doi:10.1007/s11926-018-0770-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Saung, M. T., Pelosof, L., Casak, S., Donoghue, M., Lemery, S., Yuan, M., et al. (2021). FDA Approval Summary: Nivolumab Plus Ipilimumab for the Treatment of Patients with Hepatocellular Carcinoma Previously Treated with Sorafenib. Oncol. 26 (9), 797–806. doi:10.1002/onco.13819

CrossRef Full Text | Google Scholar

Schubert, D., Bode, C., Kenefeck, R., Hou, T. Z., Wing, J. B., Kennedy, A., et al. (2014). Autosomal Dominant Immune Dysregulation Syndrome in Humans with CTLA4 Mutations. Nat. Med. 20 (12), 1410–1416. doi:10.1038/nm.3746

PubMed Abstract | CrossRef Full Text | Google Scholar

Schwabe, R. F., and Jobin, C. (2013). The Microbiome and Cancer. Nat. Rev. Cancer 13 (11), 800–812. doi:10.1038/nrc3610

PubMed Abstract | CrossRef Full Text | Google Scholar

Sender, R., Fuchs, S., and Milo, R. (2016). Revised Estimates for the Number of Human and Bacteria Cells in the Body. Plos Biol. 14 (8), e1002533. doi:10.1371/journal.pbio.1002533

PubMed Abstract | CrossRef Full Text | Google Scholar

Seton-Rogers, S. (2021). Microbiota Links to Immunotherapy Toxicity. Nat. Rev. Cancer 21 (9), 540. doi:10.1038/s41568-021-00390-w

CrossRef Full Text | Google Scholar

Shahbaz, S., Bozorgmehr, N., Koleva, P., Namdar, A., Jovel, J., Fava, R. A., et al. (2018). CD71+VISTA+ Erythroid Cells Promote the Development and Function of Regulatory T Cells through TGF-β. Plos Biol. 16 (12), e2006649. doi:10.1371/journal.pbio.2006649

PubMed Abstract | CrossRef Full Text | Google Scholar

Sharma, P., Hu-Lieskovan, S., Wargo, J. A., and Ribas, A. (2017). Primary, Adaptive, and Acquired Resistance to Cancer Immunotherapy. Cell 168 (4), 707–723. doi:10.1016/j.cell.2017.01.017

PubMed Abstract | CrossRef Full Text | Google Scholar

Shi, L., Chen, L., Wu, C., Zhu, Y., Xu, B., Zheng, X., et al. (2016). PD-1 Blockade Boosts Radiofrequency Ablation-Elicited Adaptive Immune Responses against Tumor. Clin. Cancer Res. 22 (5), 1173–1184. doi:10.1158/1078-0432.CCR-15-1352

PubMed Abstract | CrossRef Full Text | Google Scholar

Shiratori, T., Miyatake, S., Ohno, H., Nakaseko, C., Isono, K., Bonifacino, J. S., et al. (1997). Tyrosine Phosphorylation Controls Internalization of CTLA-4 by Regulating its Interaction with Clathrin-Associated Adaptor Complex AP-2. Immunity 6 (5), 583–589. doi:10.1016/s1074-7613(00)80346-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Sierro, S., Romero, P., and Speiser, D. E. (2011). The CD4-like Molecule LAG-3, Biology and Therapeutic Applications. Expert Opin. Ther. Targets 15 (1), 91–101. doi:10.1517/14712598.2011.540563

PubMed Abstract | CrossRef Full Text | Google Scholar

Stamatouli, A. M., Quandt, Z., Perdigoto, A. L., Clark, P. L., Kluger, H., Weiss, S. A., et al. (2018). Collateral Damage: Insulin-Dependent Diabetes Induced With Checkpoint Inhibitors. Diabetes 67 (8), 1471–1480. doi:10.2337/dbi18-0002

PubMed Abstract | CrossRef Full Text | Google Scholar

Stutvoet, T. S., Kol, A., Vries, E. G., Bruyn, M., Fehrmann, R. S., Terwisscha van Scheltinga, A. G., et al. (2019). MAPK Pathway Activity Plays a Key Role in PD‐L1 Expression of Lung Adenocarcinoma Cells. J. Pathol. 249 (1), 52–64. doi:10.1002/path.5280

CrossRef Full Text | Google Scholar

Takeuchi, Y., Hirota, K., and Sakaguchi, S. (2020). Impaired T Cell Receptor Signaling and Development of T Cell-Mediated Autoimmune Arthritis. Immunol. Rev. 294 (1), 164–176. doi:10.1111/imr.12841

PubMed Abstract | CrossRef Full Text | Google Scholar

Tan, S., Xu, Y., Wang, Z., Wang, T., Du, X., Song, X., et al. (2020). Tim-3 Hampers Tumor Surveillance of Liver Resident and Conventional NK Cells by Disrupting PI3K Signaling. Cancer Res. 80 (5), canres.2332.2019–1142. doi:10.1158/0008-5472.CAN-19-2332

PubMed Abstract | CrossRef Full Text | Google Scholar

Tarhini, A. A., Kang, N., Lee, S. J., Hodi, F. S., Cohen, G. I., Hamid, O., et al. (2021). Immune Adverse Events (irAEs) with Adjuvant Ipilimumab in Melanoma, Use of Immunosuppressants and Association with Outcome: ECOG-ACRIN E1609 Study Analysis. J. Immunother. Cancer 9 (5), e002535. doi:10.1136/jitc-2021-002535

CrossRef Full Text | Google Scholar

Tavares, A. B. M. L. A., Lima Neto, J. X., Fulco, U. L., and Albuquerque, E. L. (2018). Inhibition of the Checkpoint Protein PD-1 by the Therapeutic Antibody Pembrolizumab Outlined by Quantum Chemistry. Sci. Rep. 8 (1), 1840. doi:10.1038/s41598-018-20325-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Tekguc, M., Wing, J. B., Osaki, M., Long, J., and Sakaguchi, S. (2021). Treg-expressed CTLA-4 Depletes CD80/CD86 by Trogocytosis, Releasing Free PD-L1 on Antigen-Presenting Cells. Proc. Natl. Acad. Sci. USA 118 (30), e2023739118. doi:10.1073/pnas.2023739118

PubMed Abstract | CrossRef Full Text | Google Scholar

Triebel, F., Jitsukawa, S., Baixeras, E., Roman-Roman, S., Genevee, C., Viegas-Pequignot, E., et al. (1990). LAG-3, a Novel Lymphocyte Activation Gene Closely Related to CD4. J. Exp. Med. 171 (5), 1393–1405. doi:10.1084/jem.171.5.1393

CrossRef Full Text | Google Scholar

Twyman-Saint Victor, C., Rech, A. J., Maity, A., Rengan, R., Pauken, K. E., Stelekati, E., et al. (2015). Radiation and Dual Checkpoint Blockade Activate Non-redundant Immune Mechanisms in Cancer. Nature 520 (7547), 373–377. doi:10.1038/nature14292

PubMed Abstract | CrossRef Full Text | Google Scholar

Urban-Wojciuk, Z., Khan, M. M., Oyler, B. L., Fåhraeus, R., Marek-Trzonkowska, N., Nita-Lazar, A., et al. (2019). The Role of TLRs in Anti-cancer Immunity and Tumor Rejection. Front. Immunol. 10, 2388. doi:10.3389/fimmu.2019.02388

PubMed Abstract | CrossRef Full Text | Google Scholar

Vaddepally, R. K., Kharel, P., Pandey, R., Garje, R., and Chandra, A. B. (2020). Review of Indications of FDA-Approved Immune Checkpoint Inhibitors Per NCCN Guidelines with the Level of Evidence. Cancers 12 (3), 738. doi:10.3390/cancers12030738

PubMed Abstract | CrossRef Full Text | Google Scholar

van de Weyer, P. S., Muehlfeit, M., Klose, C., Bonventre, J. V., Walz, G., and Kuehn, E. W. (2006). A Highly Conserved Tyrosine of Tim-3 Is Phosphorylated upon Stimulation by its Ligand Galectin-9. Biochem. Biophysical Res. Commun. 351 (2), 571–576. doi:10.1016/j.bbrc.2006.10.079

CrossRef Full Text | Google Scholar

van der Merwe, P. A., Bodian, D. L., Daenke, S., Linsley, P., and Davis, S. J. (1997). CD80 (B7-1) Binds Both CD28 and CTLA-4 with a Low Affinity and Very Fast Kinetics. J. Exp. Med. 185 (3), 393–404. doi:10.1084/jem.185.3.393

CrossRef Full Text | Google Scholar

van Elsas, A., Sutmuller, R. P. M., Hurwitz, A. A., Ziskin, J., Villasenor, J., Medema, J.-P., et al. (2001). Elucidating the Autoimmune and Antitumor Effector Mechanisms of a Treatment Based on Cytotoxic T Lymphocyte Antigen-4 Blockade in Combination with a B16 Melanoma Vaccine. J. Exp. Med. 194 (4), 481–490. doi:10.1084/jem.194.4.481

CrossRef Full Text | Google Scholar

Vance, R. E., Eichberg, M. J., Portnoy, D. A., and Raulet, D. H. (2017). Listening to Each Other: Infectious Disease and Cancer Immunology. Sci. Immunol. 2 (7), eaai9339. doi:10.1126/sciimmunol.aai9339

PubMed Abstract | CrossRef Full Text | Google Scholar

Vétizou, M., Pitt, J. M., Daillère, R., Lepage, P., Waldschmitt, N., Flament, C., et al. (2015). Anticancer Immunotherapy by CTLA-4 Blockade Relies on the Gut Microbiota. Science 350 (6264), 1079–1084. doi:10.1126/science.aad1329

PubMed Abstract | CrossRef Full Text | Google Scholar

Walker, L. S. K. (2017). EFIS Lecture: Understanding the CTLA-4 Checkpoint in the Maintenance of Immune Homeostasis. Immunol. Lett. 184, 43–50. doi:10.1016/j.imlet.2017.02.007

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, J., Sanmamed, M. F., Datar, I., Su, T. T., Ji, L., Sun, J., et al. (2019a). Fibrinogen-like Protein 1 Is a Major Immune Inhibitory Ligand of LAG-3. Cell 176 (1-2), 334–347. doi:10.1016/j.cell.2018.11.010

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, J., Wu, G., Manick, B., Hernandez, V., Renelt, M., Erickson, C., et al. (2019b). VSIG-3 as a Ligand of VISTA Inhibits Human T-Cell Function. Immunology 156 (1), 74–85. doi:10.1111/imm.13001

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, L., Le Mercier, I., Putra, J., Chen, W., Liu, J., Schenk, A. D., et al. (2014). Disruption of the Immune-Checkpoint VISTA Gene Imparts a Proinflammatory Phenotype with Predisposition to the Development of Autoimmunity. Proc. Natl. Acad. Sci. 111 (41), 14846–14851. doi:10.1073/pnas.1407447111

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, L., Rubinstein, R., Lines, J. L., Wasiuk, A., Ahonen, C., Guo, Y., et al. (2011). VISTA, a Novel Mouse Ig Superfamily Ligand that Negatively Regulates T Cell Responses. J. Exp. Med. 208 (3), 577–592. doi:10.1084/jem.20100619

CrossRef Full Text | Google Scholar

Wang, Y., and Li, G. (2019). PD-1/PD-L1 Blockade in Cervical Cancer: Current Studies and Perspectives. Front. Med. 13 (4), 438–450. doi:10.1007/s11684-018-0674-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, Y., Wiesnoski, D. H., Helmink, B. A., Gopalakrishnan, V., Choi, K., DuPont, H. L., et al. (2018). Fecal Microbiota Transplantation for Refractory Immune Checkpoint Inhibitor-Associated Colitis. Nat. Med. 24 (12), 1804–1808. doi:10.1038/s41591-018-0238-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang-Gillam, A., Plambeck-Suess, S., Goedegebuure, P., Simon, P. O., Mitchem, J. B., Hornick, J. R., et al. (2013). A Phase I Study of IMP321 and Gemcitabine as the Front-Line Therapy in Patients with Advanced Pancreatic Adenocarcinoma. Invest. New Drugs 31 (3), 707–713. doi:10.1007/s10637-012-9866-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Weiss, S. A., and Sznol, M. (2021). Resistance Mechanisms to Checkpoint Inhibitors. Curr. Opin. Immunol. 69, 47–55. doi:10.1016/j.coi.2021.02.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Wolf, Y., Anderson, A. C., and Kuchroo, V. K. (2020). TIM3 Comes of Age as an Inhibitory Receptor. Nat. Rev. Immunol. 20 (3), 173–185. doi:10.1038/s41577-019-0224-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, Y., Sang, M., Liu, F., Zhang, J., Li, W., Li, Z., et al. (2020). Epigenetic Modulation Combined with PD-1/pd-L1 Blockade Enhances Immunotherapy Based on MAGE-A11 Antigen-specific CD8+T Cells against Esophageal Carcinoma. Carcinogenesis 41 (7), 894–903. doi:10.1093/carcin/bgaa057

PubMed Abstract | CrossRef Full Text | Google Scholar

Wykes, M. N., and Lewin, S. R. (2018). Immune Checkpoint Blockade in Infectious Diseases. Nat. Rev. Immunol. 18 (2), 91–104. doi:10.1038/nri.2017.112

PubMed Abstract | CrossRef Full Text | Google Scholar

Xu, C., Chen, Y.-P., Du, X.-J., Liu, J.-Q., Huang, C.-L., Chen, L., et al. (2018). Comparative Safety of Immune Checkpoint Inhibitors in Cancer: Systematic Review and Network Meta-Analysis. BMJ 363, k4226. doi:10.1136/bmj.k4226

PubMed Abstract | CrossRef Full Text | Google Scholar

Xu, F., Liu, J., Liu, D., Liu, B., Wang, M., Hu, Z., et al. (2014). LSECtin Expressed on Melanoma Cells Promotes Tumor Progression by Inhibiting Antitumor T-Cell Responses. Cancer Res. 74 (13), 3418–3428. doi:10.1158/0008-5472.CAN-13-2690

PubMed Abstract | CrossRef Full Text | Google Scholar

Yang, X., Jiang, X., Chen, G., Xiao, Y., Geng, S., Kang, C., et al. (2013). T Cell Ig Mucin-3 Promotes Homeostasis of Sepsis by Negatively Regulating the TLR Response. J.I. 190 (5), 2068–2079. doi:10.4049/jimmunol.1202661

CrossRef Full Text | Google Scholar

Zhang, Y., Ma, C. J., Wang, J. M., Ji, X. J., Wu, X. Y., Moorman, J. P., et al. (2012). Tim-3 Regulates Pro- and Anti-inflammatory Cytokine Expression in Human CD14+ Monocytes. J. Leukoc. Biol. 91 (2), 189–196. doi:10.1189/jlb.1010591

CrossRef Full Text | Google Scholar

Zhu, C., Anderson, A. C., Schubart, A., Xiong, H., Imitola, J., Khoury, S. J., et al. (2005). The Tim-3 Ligand Galectin-9 Negatively Regulates T Helper Type 1 Immunity. Nat. Immunol. 6 (12), 1245–1252. doi:10.1038/ni1271

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhu, C., Sakuishi, K., Xiao, S., Sun, Z., Zaghouani, S., Gu, G., et al. (2015). Erratum: Corrigendum: An IL-27/NFIL3 Signalling axis Drives Tim-3 and IL-10 Expression and T-Cell Dysfunction. Nat. Commun. 6, 7657. doi:10.1038/ncomms8657

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: immune checkpoint, immunotherapy, cancer, microbiome, PD-1/PD-L1

Citation: Cai X, Zhan H, Ye Y, Yang J, Zhang M, Li J and Zhuang Y (2021) Current Progress and Future Perspectives of Immune Checkpoint in Cancer and Infectious Diseases. Front. Genet. 12:785153. doi: 10.3389/fgene.2021.785153

Received: 28 September 2021; Accepted: 03 November 2021;
Published: 30 November 2021.

Edited by:

Tao Huang, Shanghai Institute of Nutrition and Health (CAS), China

Reviewed by:

Guomin Shen, Henan University of Science and Technology, China
Bin Yi, Louisiana State University, United States

Copyright © 2021 Cai, Zhan, Ye, Yang, Zhang, Li and Zhuang. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Yuan Zhuang, zhuangyuan723@163.com; Jing Li, lijing2020@hrbmu.edu.cn; Minghui Zhang, cfzhangminghui@163.com

These authors have contributed equally to this work

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.